US10500288B2 - Cytotoxic HEXIM1 peptides and uses thereof - Google Patents

Cytotoxic HEXIM1 peptides and uses thereof Download PDF

Info

Publication number
US10500288B2
US10500288B2 US15/555,482 US201615555482A US10500288B2 US 10500288 B2 US10500288 B2 US 10500288B2 US 201615555482 A US201615555482 A US 201615555482A US 10500288 B2 US10500288 B2 US 10500288B2
Authority
US
United States
Prior art keywords
peptide
seq
cells
peptides
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
US15/555,482
Other versions
US20180243441A1 (en
Inventor
Sheng-Hao Chao
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Agency for Science Technology and Research Singapore
Original Assignee
Agency for Science Technology and Research Singapore
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Agency for Science Technology and Research Singapore filed Critical Agency for Science Technology and Research Singapore
Assigned to AGENCY FOR SCIENCE, TECHNOLOGY AND RESEARCH reassignment AGENCY FOR SCIENCE, TECHNOLOGY AND RESEARCH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHAO, SHENG-HAO
Publication of US20180243441A1 publication Critical patent/US20180243441A1/en
Application granted granted Critical
Publication of US10500288B2 publication Critical patent/US10500288B2/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • A61K47/6931Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/66Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid the modifying agent being a pre-targeting system involving a peptide or protein for targeting specific cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/572Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca

Definitions

  • the present invention generally relates to the field of biochemistry.
  • the present invention relates to cytotoxic peptides, compositions containing cytotoxic peptides and the use of the peptides and compositions in the prevention and treatment of cancers.
  • Cancer is the second leading cause of death worldwide, accounting for 8.2 million deaths in 2012. It is expected that annual cancer cases will rise from 14 million in 2012 to 22 million within the next two decades.
  • the present invention refers to an isolated cytotoxic peptide having sequence identity of at least 66% to SEQ ID NO: 3 or functional part thereof.
  • the present invention refers to an isolated cytotoxic peptide comprising at least one amino acid sequence represented by the consensus sequence Q 1 -L 2 -G 3 -X 4 -X 5 -X 6 -X 7 -X 8 -X 9 -R 10 -P 11 -S 12 -X 13 -X 14 -X 15 -R 16 -H 17 -W 18 (SEQ ID NO: 36), or functional part thereof, wherein X is any amino acid.
  • the present invention refers to an isolated nucleic acid molecule comprising the nucleotide sequence encoding the isolated cytotoxic peptide as described herein.
  • the present invention refers to a vector comprising the isolated nucleic acid molecule as described herein.
  • the present invention refers to a pharmaceutical composition
  • a pharmaceutical composition comprising the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, or the vector as described herein.
  • the present invention refers to a method of treating or preventing cancer in a subject, comprising administering the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, the vector or the pharmaceutical composition as described herein.
  • the present invention refers to a method of eliminating undifferentiated stem cells in a subject having stem cell transplantation, comprising administering the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, the vector or the pharmaceutical composition as described herein.
  • the present invention refers to a method of treating or preventing obesity in a subject, comprising administering the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, the vector or the pharmaceutical composition as described herein.
  • FIG. 1 shows FGF-tagged BR peptide (SEQ ID NO: 19) and FGF-tagged mutant BR peptides induce cell death in various cell lines.
  • FGF tag is a cell membrane-translocating peptide derived from Kaposi fibroblast growth factor.
  • A Alignment of the HDM2 ubiquitination sites of p53 (amino acids (AAs) 370-386, SEQ ID NO: 34) and HEXIM1 (AAs 150-161, SEQ ID NO: 35) shows that ubiquitin sites of p53 and HEXIM1 exhibit similar distribution of the lysine residues (the HDM2-ubiquitinated lysine residues are underlined).
  • HCT116 (p53 WT) and HCT116 (p53 KO) cells were treated with FGF-X13, FGF-BR, FGF-BR-RRR12 (SEQ ID NO: 20) and FGF-BR-ILAA (SEQ ID NO: 21) peptides at various concentrations overnight before cell viability assays were performed. Data representative of at least three independent experiments performed in triplicate were shown with values expressed as mean ⁇ SD. The results indicate that FGF-tagged BR peptide shows similar level of cytotoxicity in HCT116 p53 wild-type (p53 WT) and HCT116 p53 knockout (p53 KO) cell lines. FGF-tagged mutant BR peptides, BR(RRR12) and BR(ILAA) also show cytotoxicity in both HCT116 (p53 WT) and HCT116 (p53KO) cell lines.
  • FIG. 2 shows FGF-/LTV-tagged BR peptide induced rapid cell death independently of apoptosis.
  • A shows representative time-lapse differential interference contrast (DIC) snapshots of treated HCT116 (p53 WT) and HCT116 (p53 KO) cells (bar represents 100 ⁇ m).
  • HCT116 (p53 WT) and HCT116 (p53 KO) cells were cultured on glass slides overnight, treated with vehicle control (0.5% dimethyl sulfoxide (DMSO)) or the indicated FGF peptides (30 ⁇ M).
  • DMSO dimethyl sulfoxide
  • MCF7 cells were incubated with or without z-VAD-Fmk (for 90 min) and then with indicated LTV-tagged peptides (30 ⁇ M) for thirty minutes at 37° C.
  • Treated cells were subjected to the cytotoxicity assay as described in Example 9—Materials and Methods. Data representative of at least three independent experiments performed in triplicate were shown with values expressed as mean ⁇ SD. The results indicate that LTV-tagged BR (SEQ ID NO: 15) has significant cytotoxic effect on MCF7 cell line, and the effect could not be inhibited by z-VAD-Fmk.
  • FIG. 3 shows FGF-tagged BR peptide decreases mitochondrial membrane potential in HCT116 (p53 WT) cell line but not in HCT116 (p53 KO) cell line.
  • A shows bar chart of mitochondrial membrane potential in HCT116 (p53 WT) cell line treated with negative and positive controls and the FGF-tagged BR peptide.
  • B shows bar chart of mitochondrial membrane potential in HCT116 (p53 KO) cell line treated with negative and positive controls and the FGF-tagged BR peptide.
  • HCT116 (p53 WT) and HCT116 (p53 KO) cells were cultured in 96-well plates, treated with indicated FGF-tagged peptides (30 ⁇ M) for three minutes.
  • Treated cells were subjected to mitochondrial membrane potential (MMP) measurement using JC-1 fluorescence-based assay for six independent experiments. Results were summarized as mean ⁇ SD (***, P ⁇ 0.0001; ns: not significant, Student's t test). The results indicate that FGF-tagged BR peptide decreased mitochondrial membrane potential in HCT116 cell line is dependent on p53.
  • FIG. 4 shows FGF-tagged BR peptide alters the sub-cellular localization and protein level of endogenous NPM.
  • A representative immunofluorescence images showing the sub-cellular localization of ⁇ -NPM (bar represents 10 ⁇ m).
  • HCT116 (p53 WT) and HCT116 (p53KO) cells were cultured on glass slides overnight, treated with FGF-X13 or FGF-BR (30 ⁇ M). Cells treated with FGF-X13 peptide or vehicle, DMSO (0.5%), was used as controls.
  • Treated cells were immunostained with an anti-NPM antibody and analyzed by laser scanning confocal microscopy (Zeiss).
  • NPM protein was quantified as described in Example 9—Materials and Methods. The results show that endogenous level of NPM is reduced in cells treated with FGF-tagged BR peptide, in both HCT116 (p53 WT) and HCT116 (p53 KO) cell lines.
  • FIG. 5 Cell viability plots showing the effects of LTV-tagged BR peptide and variants thereof, as well as untagged BR peptide and variants thereof, in two breast cancer cell lines, (A) MCF7 and (B) MDA-MB-231.
  • Cells were treated with indicated LTV-tagged peptides at different concentrations overnight before cell viability assays were performed.
  • Both LTV-tagged BR and LTV-tagged BR (RRR12) peptides decreased cell viability in (A) MCF7 and (B) MDA-MB-231 cell lines. Effects of the indicated un-tagged peptides on the viability of (C) MCF7 and (D) MDA-MB-231 cells were analyzed by cell viability assays.
  • the untagged X13 was used as a negative control.
  • the results indicate that untagged BR and BR (RRR12) peptides have no effect on cell viability in (C) MCF7 and (D) MDA-MB-231 cell lines. Data representative of at least three independent experiments performed in triplicate were shown with values expressed as mean ⁇ SD. The results indicate that LTV-tagged BR peptide decreases cell viability in breast cancer cells, while BR peptide alone shows no effect on cell viability.
  • FIG. 6 shows the untagged HEXIM1 BR peptide fails to enter cells by itself, while another cytotoxic peptide, KLA, can enter the cell without a cell penetrating and/or cell targeting tag.
  • A shows images of confocal microscopy of MCF7 cells treated with vehicle control and the indicated peptides (bar represents 10 ⁇ m). MCF7 cells were cultured on glass slides overnight before incubation with the indicated FITC-labeled peptides (30 ⁇ M) for 30 min and subsequently analyzed by laser scanning confocal microscopy. Nuclei were visualized by DAPI. Cells treated with vehicle, DMSO (0.5%) were used as control. Pointed arrow indicates FITC fluorescence.
  • results show that no FITC fluorescence can be defected in cells treated with BR-FITC peptide, while FITC fluorescence can be detected in cells treated with LTC-BR-FITC, KLA-FITC and LTV-KLA-FITC peptides.
  • B shows bar chart of FITC-positive cells quantified by flow cytometry. Data represent percentage of fluorescence-positive cells in total cell population. Results were summarized as mean ⁇ SD of three independent experiments (***, P ⁇ 0.0001, Student's t test). The results indicate that no significant cellular uptake of BR-FITC was observed, while the cellular uptake of KLA-FITC, LTV-BR-FITC and LTV-KLA-FITC is significantly higher compared to BR-FITC.
  • FIG. 7 shows cell viability plots (upper panel) of cells treated control, LTV-tagged BR and mutated variants thereof (sequences indicated in the lower panel).
  • MDA-MB-231 breast cancer cells were treated with indicated truncated LTV-tagged peptides at various concentrations overnight before cell viability assays were performed.
  • Cells treated with LTV-X13 were used as negative control, cells treated with LTV-BR were used as positive control.
  • Data representative of at least three independent experiments performed in triplicate were shown with values expressed as mean ⁇ SD. The results indicate that LTV-tagged BR peptide requires all three stretches of basic residues (KKK, RRR and KKK) to establish full potency in cell killing.
  • FIG. 8 shows cell viability plots (upper panel) of cells treated control, LTV-tagged BR and alanine substituted variants thereof (sequences indicated in the lower panel).
  • MDA-MB-231 breast cancer cells were treated with indicated alanine-substituted LTV-tagged peptides at 10 ⁇ M and 30 ⁇ M overnight before cell viability assays were performed.
  • Cells treated with LTV-X13 were used as negative control, cells treated with LTV-BR were used as positive control.
  • Data representative of at least three independent experiments performed in triplicate were shown with values expressed as mean ⁇ SD. The results indicate that alanine residue substitution in the middle and C-terminal regions of the HEXIM1 BR peptide abolished the cytotoxic activity of LTV-BR.
  • FIG. 9 shows cell viability plots of various cell lines treated with control and LTV-tagged BR and LTV-tagged KLA peptides.
  • Chinese hamster ovary cancer cell line, CHO-K1; multiple myeloma cell line, OPM-2, human foreskin fibroblasts, HFF; and human lung fibroblasts, WI-38 were treated with indicated LTV-fused peptides at various concentrations overnight before cell viability assays were performed.
  • Cells treated with LTV-X13 was used as a negative control.
  • Data representative of at least three independent experiments performed in triplicate were shown with values expressed as mean ⁇ SD. The results indicate that LTV-tagged KLA peptide exhibits non-specific killing on non-breast cancer cell lines and normal human fibroblasts, while LTV-tagged BR peptide does not show such non-specific cytotoxicity.
  • FIG. 10 shows cell viability plots of (A) undifferentiated 3T3 cells and (B) adipose cells treated with BR D and KLA D peptides tagged with lipocyte-targeting peptide, ATS (ATS-BR D and ATS-KLA D peptides).
  • ATS-BR D and ATS-KLA D peptides The results indicate that the ATS-BR D peptide has no cytotoxic effect on undifferentiated 3T3 mouse fibroblast cells, but has significant cytotoxic effect on differentiated adipose cells.
  • the cytotoxic effect of ATS-BR D peptide on adipose cells is of similar potency as ATS-KLA D peptide.
  • FIG. 11 shows that the cytotoxic effect exerted by BR is independent of p53.
  • AML2 and AML3 cells were treated with FGF-tagged p53 and FGF-tagged BR peptides at the indicated concentrations. Cell viability was measured 16 hours post-incubation by Cell-Titer Glo. Cells treated with vehicle, DMSO (0.5%), were used as control.
  • A Cell viability plots showing the effects of FGF-tagged p53 and FGF-tagged BR peptides in AML2 cell line.
  • HEXIM1 hexamethylene bisacetamide inducible protein 1
  • an isolated cytotoxic peptide having sequence identity of at least 66% to SEQ ID NO: 3, or functional part thereof.
  • the isolated cytotoxic peptide has sequence identity of at least 72%, at least 77%, at least 83%, at least 88% or at least 94% to SEQ ID NO: 3, or functional part thereof.
  • the isolated cytotoxic peptide has sequence identity of 66% to 78%, 77% to 89%, 88% to 95% to SEQ ID NO: 3, or functional part thereof.
  • the basic region (BR) of HEXIM refers to the region between AAs 150-180 of HEXIM1, which includes a stretch of basic residues.
  • the BR is represented by the sequence of KKKHRRRPSKKKRHWKPYYKLTWEEKKKFDE (SEQ ID NO: 2).
  • an “isolated” peptide is intended to mean a peptide removed from its native environment, such as recombinantly produced peptides expressed in host cells and native or recombinant peptides which have been substantially purified by any suitable technique. Isolated peptides according to the present disclosure further include such compounds produced synthetically.
  • peptide refers to polymers of amino acid residues of any length.
  • the polymer may be linear or branched, it may comprise modified amino acids or amino acid analogues, and it may be interrupted by chemical moieties other than amino acids.
  • the terms also encompass an amino acid polymer that has been modified naturally or by intervention, for example disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labelling or bioactive component.
  • the term peptide encompasses two or more naturally occurring or synthetic amino acids linked by a covalent bond (for example, an amide bond).
  • the peptide as disclosed herein can be of various lengths, such as having 6 to 200, 8 to 190, 10 to 180, 12 to 170, 14 to 160, 16 to 150, 18 to 140, 20 to 130, 22 to 120, 24 to 110, 26 to 100, 28 to 90, 30 to 80, 32 to 70, 34 to 60, 36 to 50 or 38 to 45 amino acid residues.
  • amino acid is defined as having at least one primary, secondary, tertiary or quaternary amino group, and at least one acid group, wherein the acid group may be a carboxylic, sulfonic, or phosphoric acid, or mixtures thereof.
  • the amino groups may be “alpha”, “beta”, “gamma” to “omega” with respect to the acid group(s).
  • Suitable amino acids include, without limitation, both the D- and L-isomers of the 20 common naturally occurring amino acids found in peptides (for example, A, R, N, C, D, Q, E, G, H, I, L, K, M, F, P, S, T, W, Y, V (as known by the one-letter or three letter abbreviations)) as well as the naturally occurring and unnaturally occurring amino acids prepared by organic synthesis or other metabolic routes.
  • sequence identity refers to a relationship between two or more peptide sequences, namely a reference sequence and a given sequence to be compared with the reference sequence. Sequence identity is determined by comparing the given sequence to the reference sequence after the sequences have been optimally aligned to produce the highest degree of sequence similarity, as determined by the match between strings of such sequences. Upon such alignment, sequence identity is ascertained on a position-by-position basis, for example, the sequences are “identical” at a particular position if at that position, the amino acid residues are identical. The total number of such position identities is then divided by the total number of amino acid residues in the reference sequence to give % sequence identity. Sequence identity can be readily calculated by known methods.
  • sequence identity Some methods used to determine the sequence identity are designed to give the largest match between the sequences tested. Methods to determine sequence identity are codified in publicly available computer programs that determine sequence identity between given sequences. These programs optimally align sequences using default gap weights in order to produce the highest level of sequence identity between the given and reference sequences. As an illustration, by a peptide or polypeptide having a given amino acid sequence having at least, for example, 66% sequence identity to a reference amino acid sequence, it is intended that the given amino acid sequence of the peptide or polypeptide is identical to the reference sequence except that the given peptide or polypeptide sequence may include up to 34 amino acid alterations per each 100 amino acids of the reference amino acid sequence.
  • a given peptide or polypeptide sequence having at least 66% sequence identity with a reference amino acid sequence up to 34% of the amino acid residues in the reference sequence may be deleted or substituted with another amino acid, or a number of amino acids up to 34% of the total number of amino acid residues in the reference sequence may be inserted into the reference sequence.
  • These alterations of the reference sequence may occur at the amino or the carboxyl terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in the one or more contiguous groups within the reference sequence.
  • residue positions that are not identical differ by conservative amino acid substitutions. However, conservative substitutions are not included as a match when determining sequence identity.
  • the isolated cytotoxic peptide QLGKKILAARPSKKKRHW (SEQ ID NO: 5) has 77.8% sequence identity with SEQ ID NO: 3; the isolated cytotoxic peptides QLGRRRHRRRPSKKKRHW (SEQ ID NO: 4) and HRRRPSKKKRHW (SEQ ID NO: 37) both have 66.7% sequence identity with SEQ ID NO: 3, the isolated cytotoxic peptide QLGAAAHRRRPSKKKRHW (SEQ ID NO: 38) has 83.3% sequence identity with SEQ ID NO: 3.
  • the isolated cytotoxic peptide as described herein has at least 12, at least 13, at least 14, at least 15, at least 16 or at least 17 amino acid residues that are the same as SEQ ID NO: 3, or functional part thereof. In some other examples, the isolated cytotoxic peptide has 12 to 14 or 15 to 17 amino acid residues that are the same as SEQ ID NO: 3, or functional part thereof.
  • cytotoxic and “cytotoxicity” as used herein refer to the effect of a compound or composition of being toxic to the cells. Effects of cytotoxicity on cells include, but are not limited to, cell swelling, cell lysis, loss of cell membrane integrity, decreased rate of metabolism, decreased cell viability, cell deaths (by inducing apoptosis or necrosis). Thus, the cytotoxic effects can be exerted via various pathways. Examples of possible pathways include, but are not limited to, apoptosis-independent pathways, positive transcription elongation factor b (P-TEFb)-independent pathway, p53-independent pathway.
  • P-TEFb positive transcription elongation factor b
  • low toxicity or “reduced toxicity” as used in the present disclosure refer to a decrease in toxicity towards non-cancerous cells and tissue relative or compared to cancerous cells and tissue.
  • the low or reduced toxicity towards non-cancerous cells and tissue is at most about 0.9 times, at most about 0.8 times, at most about 0.7 times, at most about 0.6 times, at most about 0.5 times, at most about 0.4 times, at most about 0.3 times, at most about 0.2 times, at most about 0.1 times, or at most about 0.05 times the toxicity towards cancer cells and tissue samples obtained from the same or a different subject.
  • apoptosis refers to programmed cell death, characterized by cell changes such as blebbing, cell shrinkage, nuclear fragmentation, chromatin condensation, chromosomal DNA fragmentation, and global mRNA decay.
  • the term “functional part” of a peptide or amino acid sequence refers to the part of the peptide or the amino acid sequence that produces the overall function of a peptide or amino acid sequence.
  • the functional part of a peptide or amino acid sequence comprises 6 to 17, 7 to 16, 8 to 15, 9 to 14, 10 to 13 or 11 to 12 amino acid residues of the peptide or amino acid sequence.
  • the functional part of SEQ ID NO: 3 refers to the part of SEQ ID NO: 3 that has cytotoxic effect.
  • the functional part of SEQ ID NO: 3 comprises at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, at least fifteen, at least sixteen or at least seventeen amino acid residues of the peptide or amino acid sequence.
  • the functional part of SEQ ID NO: 3 comprises at least the following stretches of amino acid residues RRR (AAs 8-10 of SEQ ID NO: 3) and KKK (AAs 13-15 of SEQ ID NO: 3).
  • RRR amino acid residues 8-10 of SEQ ID NO: 3
  • KKK AAs 13-15 of SEQ ID NO: 3
  • the isolated cytotoxic peptide as described herein comprises amino acid residues at positions corresponding to amino acids 8 to 10 and 13 to 15 of SEQ ID NO: 3.
  • Exemplary peptides include, but are not limited to, the sequences of:
  • the isolated cytotoxic peptide as described above may comprise at least one amino acid sequence that can be represented by a consensus sequence.
  • an isolated cytotoxic peptide having sequence identity of at least 66% to SEQ ID NO: 3, or functional part thereof, comprising at least one amino acid sequence represented by the consensus sequence Q 1 -L 2 -G 3 -X 4 -X 5 -X 6 -X 7 -X 8 -X 9 -R 10 -P 11 -S 12 -X 13 -X 14 -X 15 -R 16 -H 17 -W 18 (SEQ ID NO: 36), or functional part thereof.
  • the isolated cytotoxic peptide has sequence identity of at least 72%, at least 77%, at least 83%, at least 88% or at least 94% to SEQ ID NO: 3, or functional part thereof. In some other examples, the isolated cytotoxic peptide has sequence identity of 66% to 78%, 77% to 89%, 88% to 95% to SEQ ID NO: 3, or functional part thereof.
  • an isolated cytotoxic peptide comprising at least one amino acid sequence represented by the consensus sequence Q 1 -L 2 -G 3 -X 4 -X 5 -X 6 -X 7 -X 8 -X 9 -R 10 -P 11 -S 12 -X 13 -X 14 -X 15 -R 16 -H 17 -W 18 (SEQ ID NO: 36), or functional part thereof.
  • Consensus sequence refers to an amino acid sequence determined by aligning a series of multiple sequences. It defines an idealized sequence that represents the predominant choice of amino acid at each corresponding position of the multiple sequences.
  • X 4 , X 5 and X 6 can be any amino acids or may be deleted. In some examples, X 4 , X 5 and X 6 are positively charged amino acids. In some examples, X 4 , X 5 and X 6 are the same. In some other examples, at least two of X 4 , X 5 and X 6 are different from each other. In some examples, X 7 can be histidine (H) or leucine (L). In some examples, X 8 and X 9 are positively charged amino acids. In some other examples, X 8 and X 9 can be small amino acid residues including but are not limited to alanine (A).
  • A alanine
  • X 8 and X 9 are the same, while in some other examples, X 8 and X 9 are different.
  • X 13 , X 14 and X 15 are positively charged amino acids.
  • X 13 , X 14 and X 15 are the same. In some other examples, at least two of X 13 , X 14 and X 15 are different from each other.
  • the positively charged amino acids may include, but are not limited to, lysine (K), arginine (R) or histidine (H).
  • the positively charged amino acids may also include positively charged unnatural amino acids such as ornithine; 2,4-diaminobutanoic acid 2,3-diaminopropanoic acid), 3-(aminoiminomethyl)amino-alanine, 2-amino-4-(aminoiminomethyl)aminobutanoic acid, N 6 -(aminoiminomethyly) lysine, 2-amino-7-(aminoiminomethyl)aminoheptanoic acid, 2,7-diaminoheptanoic acid, 2,8-diaminooxtanoic acid, 2,9-diaminononanoic acid, 2,10-diaminodecanoic acid, 4-(aminoiminomethyl)phenylalanine or 4-(aminoiminomethyl)aminophenylalanine.
  • Exemplary peptides represented by the consensus sequence Q 1 -L 2 -G 3 -X 4 -X 5 -X 6 -X 7 -X 8 -X 9 -R 10 -P 11 -S 12 -X 13 -X 14 -X 15 -R 16 -H 17 -W 18 include, but are not limited to, the following sequences:
  • the isolated cytotoxic peptide as described herein excludes the sequence of QLGKKKHRRRPSKKKRHW (SEQ ID NO: 3).
  • the isolated cytotoxic peptide comprising the amino acid sequence represented by the consensus sequence as described above can include one or more additional amino acids at either end of the amino acid sequence represented by the consensus sequence.
  • the isolated cytotoxic peptide can include at least 1, at least 3, at least 5, at least 7, at least 9, at least 11, at least 13, at least 15, at least 17, at least 19, or 2, 4, 6, 8, 10, 12, 14, 16, 18, 20 additional amino acid residues at the N-terminal and/or the C-terminal of the amino acid sequence represented by the consensus sequence.
  • the functional part of the amino acid sequence represented by the consensus sequence comprises at least the following stretches of amino acid residues X 8 -X 9 -R 10 and X 13 X 14 X 15 .
  • Exemplary peptides comprising such functional part include, but are not limited to, the following sequences:
  • the isolated cytotoxic peptide comprising the amino acid sequence represented by the consensus sequence as described above can include more than one amino acid sequence represented by the consensus sequence.
  • the isolated cytotoxic peptide as described above includes at least 2, at least 3 or at least 4 amino acid sequences represented by the consensus sequence.
  • at least 2 or at least 3 of the amino acid sequences represented by the consensus sequence are of the same sequence.
  • all the amino acid sequences represented by the consensus sequence are different.
  • the more than one amino acid sequences represented by the consensus sequence are separated by 1 to 3, 4 to 6, 7 to 9, 10 to 12, 11 to 13, 14 to 16, 17 to 19, 20 to 22 or 23 to 25 other amino acid residues.
  • at least 2 or at least 3 of the more than one amino acid sequences represented by the consensus sequence are not separated by any other amino acid residues.
  • the isolated cytotoxic peptide as described herein can exert it cytotoxicity via the apoptosis-independent pathway, the p53-independent pathway or the p-TEFb-independent pathway.
  • the isolated cytotoxic peptide or conjugate thereof is effective against cancer resistant to p53-directed therapy, cancer resistant to the TEFb-directed pathway, or cancer resistant to therapies targeting other apoptosis-dependent pathways.
  • resistant or grammatical variants are used herein in the context of being drug-resistant, and refer to the reduction in effectiveness of a drug in curing a disease or condition.
  • Drug resistance can result from, for example, defects in a pathway or defects in element(s) in a pathway targeted by the drug.
  • a patient with a mutated or an otherwise defective p53 protein will show resistance to a drug targeting the p53-dependent pathway.
  • the isolated cytotoxic peptide as disclosed herein may contain peptide modifications.
  • modification is used herein to refer to post-translational modification in general, which are modifications that occur on a peptide usually after its translation by ribosome is complete.
  • Post-translational modification generally refers to the addition of a functional group covalently to a protein. These modifications include, but are not limited to, phosphorylation, glycosylation, sulfation, biotinylation, hydroxylation, acetylation, ubiquitination, nitrosylation, methylation, acetylation and lipidation.
  • the isolated cytotoxic peptide as described herein contains at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, or 1 to 4, 5 to 8, 9 to 12, 13 to 16, 17 to 20, 21 to 24, 25 to 28, 29 to 32, 33 to 36, 37 to 40, 41 to 44, 45 to 48 or 49 to 52 modified amino acid residues.
  • all the modified amino acid residues contain the same type of modification. In some other examples, at least some of the modified amino acid residues contain different types of modifications.
  • the isolated cytotoxic peptide comprises more than one amino acid sequence represented by the consensus sequence Q 1 -L 2 -G 3 -X 4 -X 5 -X 6 -X 7 -X 8 -X 9 -R 10 -P 11 -S 12 -X 13 -X 14 -X 15 -R 16 -H 17 -W 18 (SEQ ID NO: 36), at least two of the amino acid sequences represented by the consensus sequence contain the same amino acid modifications.
  • all the amino acid sequences represented by the consensus sequence are modified differently.
  • one amino acid residue may contain multiple modifications, such as one, two or three modifications.
  • all the modifications on the same amino acid residue are of the same type. In some other examples, at least some of the modifications on the same amino acid residue are of different types. Examples of such modification include, but are not limited to glycosylation, sulfation, phosphorylation, ubiquitination, methylation, lipidation, biotinylation, hydroxylation and acetylation.
  • glycosylation refers to the addition of a glycosyl group, usually to, but not limited to an arginine, an asparagine, a cysteine, a hydroxylysine, a serine, a threonine, a tyrosine, or a tryptophan residue, resulting in a glycoprotein.
  • a glycosyl group refers to a substituent structure obtained by removing the hemiacetal hydroxyl group from the cyclic form of a monosaccharide and, by extension, of a lower oligosaccharide.
  • sulfation refers to the addition of a sulfo group usually to a tyrosine residue.
  • a sulfo group refers to group SO 3 H ⁇ , derived from sulfuric acid.
  • phosphorylation refers to the addition of a phosphate group. Protein phosphorylation commonly takes place at the serine, threonine, tyrosine, arginine, lysine, or cysteine residues. Phosphorylation may alter the structural conformation of a protein, causing it to become activated, deactivated, or modifying its function.
  • ubiquitination refers to the addition of a ubiquitin.
  • Ubiquitin is a small protein that is found in almost all cellular tissues in humans and other eukaryotic organisms, which helps to regulate the processes of other proteins in the body.
  • the genes in the human genome that produce ubiquitin include, but are not limited to: UBB, UBC, UBA52 and RPS27A.
  • Ubiquitination may affect, for example, the activity and location of a protein, as well the interaction of the ubiquitinated protein with other proteins.
  • ubiquitination may lead to the degradation of the protein.
  • the protein is degraded via the proteasome.
  • methylation refers to the addition of a methyl group.
  • a methyl group refers to an alkyl derived from methane, containing one carbon atom bonded to three hydrogen atoms, that is —CH 3 .
  • Protein methylation can commonly take place at the arginine or lysine amino acid residues.
  • lipidation refers to the addition of a lipid group.
  • examples of lipidation include but are not limited to N-Myristoylation, Palmitoylation, Glycosylphosphatidylinositol (GPI)-anchor addition and Prenylation.
  • biotinylation refers to the addition of a biotin.
  • a biotin refers to 5-[(3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl]pentanoic acid, with the following structural formula:
  • hydroxylation refers to the addition of a hydroxyl group (—OH).
  • acetylation refers to the addition of an acetyl group.
  • An acetyl group contains a methyl group single-bonded to a carbonyl, represented by the following formula:
  • amino acid changes in the polypeptide or fragment thereof may be effected by techniques well known to persons skilled in the relevant art.
  • amino acid changes may be effected by nucleotide replacement techniques which include the addition, deletion or substitution of nucleotides, under the proviso that the proper reading frame is maintained.
  • Exemplary techniques include random mutagenesis, site-directed mutagenesis, oligonucleotide-mediated or polynucleotide-mediated mutagenesis, deletion of selected region(s) through the use of existing or engineered restriction enzyme sites, and the polymerase chain reaction.
  • the cell internalization of the cytotoxic peptide disclosed herein may require facilitators such as cell penetrating or cell targeting peptides or proteins.
  • the isolated cytotoxic peptide as described herein is conjugated to at least one internalizing peptide or protein.
  • conjugate refers to the attachment of the isolated cytotoxic peptide to another object, such as other peptide(s) or polypeptide(s), protein(s), antibody(ies), or particle(s).
  • the attachments can be resulted from covalent bonding or weak interactions.
  • conjugation should be construed accordingly.
  • the isolated cytotoxic peptide as described herein can be conjugated to an antibody.
  • antibody is used in the broadest sense to refer to an immunoglobulin molecule, a fragment of an immunoglobulin molecule, or a derivative of either thereof.
  • the antibody can be a monoclonal antibody or a polyclonal antibody.
  • the antibody is an antibody for cancer therapy.
  • Examples of an antibody for cancer therapy include, but are not limited to, Ado-trastuzumab emtansine (CAS No. 1018448-65-1), Alemtuzumab (CAS No. 216503-57-0), Bevacizumab (CAS No. 216974-75-3), Blinatumomab (CAS No. 853426-35-4), Brentuximab vedotin (CAS No.
  • the isolated cytotoxic peptide as described herein can be conjugated to a protein or an antibody by, for example but not limited to, a cross-linker, a bifunctional linker or a trifunctional linker.
  • Linker groups can be, for example but not limited to, small organic compounds or peptides substituted with chemical linkers.
  • the chemical linkers can be, but not limited to, any one of the following: succinimidyl 3-(2-pyridyldithio)propionate (SPDP), succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC), succinimidyl 4-(p-maleimidophenyl)butyrate (SMPB), 4-succinimidyloxycarbonyl-alpha-methyl- ⁇ (2-pyridyldithio)toluene (SMPT), EMCS (N- ⁇ -malemidocaproyl-oxysuccinimide ester) and Sulfo-MBS (m-maleimidobenzoyl-N-hydroxysulfosuccinimide ester).
  • SPDP succinimidyl 3-(2-pyridyldithio)propionate
  • SMCC succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate
  • a thiol group on the peptide is required for conjugating the isolated cytotoxic peptide as described herein to a linker.
  • the isolated cytotoxic peptide will have at least one additional cysteine residue at either end of the peptide. Therefore, in some examples, the isolated cytotoxic peptide as described herein further comprises at least one cysteine residue at the N-terminal and/or the C-terminal end.
  • the isolated cytotoxic peptide as described herein can exert its cytotoxic effect on any cell type, once the isolated cytotoxic peptide is internalized into the cell. Therefore, depending on the type of internalizing peptide or protein being conjugated to, the isolated cytotoxic peptide as described herein can exert its cytotoxic effect on a specific type of cell.
  • the internalizing peptide or protein is a cell penetrating peptide or protein.
  • the internalizing peptide or protein is a cell targeting peptide or protein.
  • a cell penetrating peptide or protein can also function as a cell targeting peptide or protein.
  • Examples of peptides that can function as both a cell penetrating peptide and a cell targeting peptide include but are not limited to, LTVSPWY peptide (LTV, SEQ ID NO: 8), HLYVSPW peptide (Pep2, SEQ ID NO: 795), CGFYWLRSC peptide (NRP, SEQ ID NO: 796) and CQDGRMGFC peptide (BLA, SEQ ID NO: 797).
  • the cell targeting peptide or protein is a cancer cell targeting peptide or protein.
  • the cell targeting peptide or protein is a lipocyte targeting peptide or protein.
  • the cell targeting peptide or protein is a stem cell targeting peptide or protein.
  • such conjugation can result in the internationalization of both the cytotoxic peptide and the internalizing peptide or protein.
  • a cell-penetrating peptide include, but are not limited to, transactivator of transcription (TAT), penetratin, R6-Pen, transportan, MPG peptide, sweet arrow peptide (SAP), peptide from vascular endothelial-cadherin protein (pVEC), Pep-1 (KETWWETWWTEWSQPKKKRKV)(SEQ ID NO: 6), polylysines, polyarginines, model amphipathic peptide (MAP), FGF (AAVALLPAVLLALLAP) (SEQ ID NO: 798) and R6W3 (RRWWRRWRR)(SEQ ID NO: 7).
  • TAT transactivator of transcription
  • R6-Pen transportan
  • MPG peptide MPG peptide
  • Examples of a cancer cell-targeting peptide include but are not limited toarginine-glycine-aspartic acid (RGD), asparagine-glycine-arginine (NGR), TCP-1 phage peptide (TCP-1), and the peptides listed in Table 1.
  • Examples of a lipocyte-targeting peptide include but are not limited to, CKGGRAKDC peptide (ATS, SEQ ID NO: 39). Examples of stem cell targeting peptide are listed in Table 2. Examples of conjugated peptides and their effects are discussed in details in Examples 6 to 8.
  • the isolated cytotoxic peptide or conjugate thereof as described herein can exert its cytotoxicity selectively.
  • the isolated cytotoxic peptide or conjugate thereof has low toxicity towards cells and tissue that are not the target of the conjugated cell penetrating peptide or cell targeting peptide.
  • the isolated cytotoxic peptide or conjugate thereof exerts its cytotoxic effects only upon internalization into a target cell.
  • the stability of the isolated cytotoxic peptide as disclosed herein can be enhanced by conjugating to a particle, in particular a small particle.
  • a particle include but are not limited to, nanoparticle or microparticle.
  • the nanoparticle or microparticle is made from materials including but not limited to, metal, silica, carbon, polymeric materials, and mixtures thereof.
  • metal include, but are not limited to, gold and silver.
  • polymeric materials include, but are not limited to, bio-degradable polymers such as a poly(lactide-co-glycolide), poly(lactic acid), poly(alkylene glycol), polybutylcyanoacrylate, poly(methylmethacrylate-co-methacrylic acid), poly-allylamine, polyanhydride and polyhydroxybutyric acid.
  • bio-degradable polymers such as a poly(lactide-co-glycolide), poly(lactic acid), poly(alkylene glycol), polybutylcyanoacrylate, poly(methylmethacrylate-co-methacrylic acid), poly-allylamine, polyanhydride and polyhydroxybutyric acid.
  • nucleic acid molecule comprising the nucleotide sequence encoding a peptide as described herein.
  • the disclosure is not limited to a specific nucleic acid molecule but includes all nucleic acid molecules comprising a nucleotide sequence coding for the peptide as described herein.
  • a nucleic acid molecule such as DNA (including cDNA and genomic DNA), RNA (such as mRNA), is regarded to be ‘capable of expressing a nucleic acid molecule or a coding nucleotide sequence’ or capable ‘to allow expression of a nucleotide sequence’ if it contains regulatory nucleotide sequences which contain transcriptional and translational information and such sequences are “operably linked” to nucleotide sequences which encode the peptide.
  • An operable linkage is a linkage in which the regulatory DNA sequences and the DNA sequences sought to be expressed are connected in such a way as to permit gene sequence expression.
  • the precise nature of the regulatory regions needed for gene sequence expression may vary from organism to organism, but shall, in general include a promoter region which, in prokaryotes, contains only the promoter or both the promoter which directs the initiation of RNA transcription as well as the DNA sequences which, when transcribed into RNA will signal the initiation of synthesis.
  • Such regions will normally include non-coding regions which are located 5′ and 3′ to the nucleotide sequence to be expressed and which are involved with initiation of transcription and translation such as the TATA box, capping sequence and CAAT sequences. These regions can for example, also contain enhancer sequences or translated signal and leader sequences for targeting the produced peptide to a specific compartment of a host cell, which is used for producing a peptide described above.
  • the isolated nucleic acid molecule as described herein can be obtained by cloning or produced synthetically. Examples of the isolated nucleic acid molecules are listed in Table 3.
  • the isolated nucleic acid molecule comprising the nucleotide sequence encoding the peptide as disclosed herein can be comprised in a vector, for example an expression vector.
  • a vector comprising an isolated nucleic acid molecule as described herein.
  • Representative vectors include plasmids, cosmids, and viral vectors.
  • Vectors can also comprise nucleic acids including expression control elements, such as transcription/translation control signals, origins of replication, polyadenylation signals, internal ribosome entry sites, promoters and enhancers, wherein the control elements are operatively associated with a nucleic acid encoding a gene product.
  • a vector can be introduced into targeting cells using any suitable method known in the art for introducing DNA into cells, including but not limited to microinjection, electroporation, calcium phosphate precipitation, liposome-mediated delivery, viral infection, protoplast fusion, and particle-mediated uptake.
  • the vector comprising the isolated nucleic acid molecule can be transformed into host cells capable of expressing the genes.
  • the transformation can be carried out in accordance with standard techniques.
  • the disclosure is also directed to a (recombinant) host cell containing a vector as described herein.
  • the transformed host cells can be cultured under conditions suitable for expression of the nucleotide sequence encoding the peptide as described above.
  • Host cells can be established, adapted and completely cultivated under serum free conditions, and optionally in media which are free of any protein/peptide of animal origin.
  • RPMI-1640 Sigma
  • Dulbecco's Modified Eagle's Medium (DMEM; Sigma)
  • MEM Minimal Essential Medium
  • CHO-S-SFMII Invitrogen
  • serum free-CHO Medium (Sigma)
  • protein-free CHO Medium (Sigma) are exemplary appropriate nutrient solutions.
  • any of the media may be supplemented as necessary with a variety of compounds, examples of which are hormones and/or other growth factors (such as insulin, transferrin, epidermal growth factor, insulin like growth factor), salts (such as sodium chloride, calcium, magnesium, phosphate), buffers (such as HEPES), nucleosides (such as adenosine, thymidine), glutamine, glucose or other equivalent energy sources, antibiotics, trace elements. Any other necessary supplements may also be included at appropriate concentrations that are known to those skilled in the art.
  • hormones and/or other growth factors such as insulin, transferrin, epidermal growth factor, insulin like growth factor
  • salts such as sodium chloride, calcium, magnesium, phosphate
  • buffers such as HEPES
  • nucleosides such as adenosine, thymidine
  • glutamine glucose or other equivalent energy sources
  • antibiotics trace elements
  • compositions for example pharmaceutical compositions, suitable for administration.
  • the peptide or conjugate thereof, the nucleic acid molecule or the vector can be administered with a pharmaceutically acceptable carrier.
  • a “carrier” can include any pharmaceutically acceptable carrier as long as the carrier is compatible with other ingredients of the formulation and not injurious to the patient. Accordingly, pharmaceutical compositions for use may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising, but not limited to, an isolated cytotoxic peptide or conjugate thereof, an isolated nucleic acid molecule or a vector as described herein.
  • the pharmaceutical composition further comprises one or more pharmaceutically acceptable excipients, vehicles or carriers.
  • Examples of pharmaceutically acceptable excipients, carriers or diluents are demineralised or distilled water; saline solution; vegetable based oils such as peanut oil, safflower oil, olive oil, cottonseed oil, maize oil, sesame oils such as peanut oil, safflower oil, olive oil, cottonseed oil, maize oil, sesame oil, arachis oil or coconut oil; silicone oils, including polysiloxanes, such as methyl polysiloxane, phenyl polysiloxane and methylphenyl polysolpoxane; volatile silicones; mineral oils such as liquid paraffin, soft paraffin or squalane; cellulose derivatives such as methyl cellulose, ethyl cellulose, carboxymethylcellulose, sodium carboxymethylcellulose or hydroxypropylmethylcellulose; lower alkanols, for example ethanol or iso-propanol; lower aralkanols; lower polyalkylene glycols or lower alkylene glycols,
  • the isolated cytotoxic peptide or conjugate thereof as described herein may be present in the compositions in any of a wide variety of forms. For example, two, three, four or more peptides or conjugates thereof may be merely mixed together or may be more closely associated through complexation, crystallization, or ionic or covalent bonding.
  • the peptides or conjugates thereof can also be formulated as prodrugs.
  • prodrug refers to compounds that rapidly convert in vivo into pharmacologically active compounds. Suitable prodrugs can be made, for instance, by conjugating the isolated cytotoxic peptide as described herein to one or more protective peptides.
  • Various linkers known in the art can be used in such conjugations, for example but not limited to valine-citrulline dipeptide linker. Such linkers can be cleavable, resulting in the release of the pharmacologically active compounds.
  • valine-citrulline dipeptide linker is cleavable by lysosomal cathepsin B, resulting in the release of the cytotoxic peptide.
  • the pharmaceutical composition further comprises one or more therapeutic agent.
  • the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule and/or the vector as described herein may be administered simultaneously, sequentially or separately from the one or more further therapeutic agent.
  • simultaneous is meant that the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule or the vector as described herein may be contained in the same pharmaceutical composition as the one or more further therapeutic agent, or they are contained in different pharmaceutical compositions but taken at the same time.
  • the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule or the vector as described herein and the one or more further therapeutic agent are contained in different pharmaceutical compositions, and are administered one immediately after another.
  • the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule or the vector as described herein and the one or more further therapeutic agent are contained in different pharmaceutical compositions, and are administered with a period of time apart. For example, they can be administered separately with about 12 hours, 10 hours, 8 hours, 6 hours, 4 hours, 2 hours, 1 hour, 30 minutes, or 15 minutes apart.
  • the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule or the vector as described herein and the one or more further therapeutic agent are contained in different pharmaceutical compositions, they can be administered using different routes.
  • the one or more therapeutic agent can be therapeutic agent against cancer, therapeutic agent for weight loss, or therapeutic agent with other effects, including but not limited to, reducing inflammation, reducing infection and relieving pain.
  • the one or more therapeutic agent is a therapeutic agent against cancer, including but not limited to, general chemotherapeutic agents such as alkylating agents, antimetabolites, anti-tumor antibiotics, topoisomerase inhibitors, mitotic inhibitors and corticosteroids, inhibitors of translation and transcription; immunotherapeutic agents such as antibodies, in particular monoclonal antibodies, cytokines and vaccines; and targeted therapeutic agents such as small molecule drugs.
  • the one or more therapeutic agent is a therapeutic agent for weight loss, including but not limited to, benzphetamine, bupropion hydrochloride, diethylpropion, lorcaserin, methamphetamine, naltrexone hydrochloride, orlistat, phendimetrazine, phentermine and sibutramine.
  • administration may be oral, intraadiposal, intraarterial, intraarticular, intracranial, intradermal, intralesional, intramuscular, intranasal, intraocularal, intrapericardial, intraperitoneal, intrapleural, intraprostatical, intrarectal, intrathecal, intratracheal, intratumoral, intraumbilical, intravenous, intravesicularl, intravitreal, liposomal, local, mucosal, enteral, parenteral, rectal, subconjunctival, subcutaneous, sublingual, topical, transbuccal, transdermal, vaginal, in crèmes, in lipid compositions, via a catheter, via a lavage, via continuous infusion, via infusion, via inhalation, via injection, via
  • compositions and formulations for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets or tablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders may be desirable.
  • compositions and formulations for parenteral, intrathecal or intraventricular administration may include sterile aqueous solutions that may also contain buffers, diluents and other suitable additives such as, but not limited to, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients.
  • compositions as described herein include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions may be generated from a variety of components that include, but are not limited to, preformed liquids, self-emulsifying solids and self-emulsifying semisolids.
  • formulations as described herein may be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredients with the pharmaceutical carrier(s) or excipient(s). In general the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • compositions as described herein may be formulated into any of many possible dosage forms including, but not limited to tablets, capsules, liquid syrups and soft gels.
  • the compositions as described herein may also be formulated as suspensions in aqueous, non-aqueous or mixed media.
  • Aqueous suspensions may further contain substances that increase the viscosity of the suspension including, for example, sodium carboxymethyl cellulose, sorbitol and/or dextran.
  • the suspension may also contain stabilizers.
  • compositions as described herein may additionally contain other adjunct components conventionally found in pharmaceutical compositions.
  • the compositions may contain additional, compatible, pharmaceutically-active materials such as, for example, antipruritics, astringents, local anesthetics or anti-inflammatory agents, or may contain additional materials useful in physically formulating various dosage forms of the compositions of the present disclosure, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
  • additional materials useful in physically formulating various dosage forms of the compositions of the present disclosure such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
  • such materials when added, should not unduly interfere with the biological activities of the components of the compositions of the present disclosure.
  • the formulations can be sterilized and, if desired, mixed with auxiliary agents, including but not limited to, lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like which do not deleteriously interact with the antibody(s) of the formulation.
  • auxiliary agents including but not limited to, lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like which do not deleteriously interact with the antibody(s) of the formulation.
  • the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, the vector or the pharmaceutical composition as described herein can be used in the treatment or prevention of diseases.
  • diseases include, but are not limited to, cancer and obesity.
  • a method of treating or preventing, for example, but not limited to cancer and obesity, in a subject comprising administering an isolated cytotoxic peptide as described herein, an isolated nucleic acid molecule as described herein, a vector as described herein or a pharmaceutical composition as described herein.
  • the major types of cancers that can be treated include but are not limited to carcinoma, sarcoma, lymphoma, germ cell tumor and blastoma.
  • the specific types of cancers that can be treated include but are not limited to breast cancer, colorectal cancer, gastric cancer, melanoma, pancreatic cancer, skin cancer, leukemia, myeloma, hepatocellular cancer, pancreatic cancer, cervical cancer, ewings tumour, neuroblastoma, kaposis sarcoma, prostate cancer, bladder cancer, melanoma, lung cancer—non small cell lung cancer (NSCLC), and small cell lung cancer (SCLC), head and neck cancer, renal cancer, lymphoma, prostate cancer, neuroblastoma, a blood cancer, testicular cancer, ovarian cancer, liver cancer or esophageal cancer, cervical cancer, non-melanoma skin cancer, glioblastoma, carcinoma, uterus cancer, chronic lymphoid leukemia, lymphoblastic
  • the cancer is invasive and/or metastatic cancer. In some other examples, the cancer is stage I cancer, stage II cancer, stage III cancer or stage IV cancer.
  • Stem cell therapy is a promising treatment for various diseases such as cancers, cardiovascular diseases, brain and spinal cord injury, blood related diseases, Parkinson's disease, Alzheimer's disease, diabetes, wound healing and so forth.
  • the major types of stem cells include but are not limited to, embryonic stem cells, induced pluripotent stem cells and tissue stem cells.
  • embryonic stem cells induced pluripotent stem cells and tissue stem cells.
  • tissue stem cells One main concern of the stem cell therapy, in particular the therapy using embryonic stem cells or induced pluripotent stem cells, is that the transplantation of any undifferentiated stem cells can lead to side effects such as the formation of teratoma, which is a tumor containing tissues derived from all three embryonic germ layers, i.e., ectoderm, mesoderm, and endoderm.
  • drugs with specific cytotoxic effect against undifferentiated stem cells can be used.
  • the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, the vector or the pharmaceutical composition as described herein can be used in the prevention or treatment of diseases associated with stem cell transplantation.
  • the stem cell transplantation can be, but not limited to, transplantation of embryonic stem cells, transplantation of induced pluripotent stem cells, or transplantation of tissue stem cells, or a mixture thereof.
  • One example of such diseases is teratoma.
  • the teratoma is benign, while in some other examples, the teratoma is malignant.
  • the subject that is being treated may be a mammal. In one specific example, the subject may be a human.
  • treatment refers to any and all uses which remedy a disease state or symptoms, prevent the establishment of disease, or otherwise prevent, hinder, retard, or reverse the progression of disease or other undesirable symptoms in any way whatsoever.
  • Treatments of cancer include but are not limited to: (i) the prevention or inhibition of cancer or cancer recurrence, (ii) the reduction or elimination of symptoms or cancer cells, and (iii) the substantial or complete elimination of the cancer in question. Treatment may be effected prophylactically (prior to the onset of the disease) or therapeutically (following diagnosis of the disease).
  • the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, the vector or the composition as described herein may be provided in an amount that is therapeutically effective.
  • the suitable amount includes a sufficient but non-toxic amount of the compound as described herein to provide the desired therapeutic effect.
  • the exact amount required will vary from subject to subject depending on factors such as the species being treated, the age and general condition of the subject, the severity of the condition being treated, the particular agent being administered, the mode of administration, and so forth, and may be determined by one of ordinary skill in the art using only routine experimentation.
  • Dosing is dependent on severity and responsiveness of the disease state to be treated, with the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved.
  • Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient. The administering physician can easily determine optimum dosages, dosing methodologies and repetition rates. Optimum dosages may vary depending on the relative potency of the composition.
  • dosage is from 0.01 ⁇ g to 100 g/kg of body weight, such as 0.01 g to 100 g, 0.05 ⁇ g to 90 g, 0.1 ⁇ g to 80 g, 0.5 ⁇ g to 70 g, 1 ⁇ g to 60 g, 10 ⁇ g to 50 g, 20 ⁇ g to 40 g, 30 ⁇ g to 30 g, 40 ⁇ g to 20 g, 50 ⁇ g to 10 g, 75 ⁇ g to 5 g, 100 ⁇ g to 4 g, 200 ⁇ g to 3 g, 300 ⁇ g to 2 g, 400 ⁇ g to 1 g, 500 ⁇ g to 900 mg, 600 ⁇ g to 800 mg, 700 ⁇ g to 700 mg, 800 ⁇ g to 600 mg, 900 ⁇ g to 500 mg, 1 mg to 400 mg, 2 mg to 300 mg, 3 mg to 200 mg, 3 mg to 100 mg, 4 mg to 90 mg, 5 mg to 80 mg, 6 mg to 70 mg, 7 mg to 60 mg, 8 mg to 50
  • the treating physician can estimate repetition rates for dosing based on measured residence times and concentrations of the drug in bodily fluids or tissues.
  • Example 1 HEXIM1 BR Peptide Induces Cell Death in a P-TEFb- and p53-Independent Fashion
  • HEXIM1 was originally identified from vascular smooth muscle cells treated with HMBA, an anti-proliferation compound.
  • HEXIM1 is known as the inhibitor of P-TEFb.
  • P-TEFb is a protein complex, which is composed of cyclin-dependent kinase 9 (CDK9) and a cyclin partner (cyclin T1, T2a, T2b, or K) with cyclin T1 being the predominant CDK9-associated cyclin.
  • CDK9 cyclin-dependent kinase 9
  • cyclin T1, T2a, T2b, or K cyclin partner
  • P-TEFb controls the elongation phase of RNAPII transcription and is required for transcriptional regulation of human immunodeficiency virus.
  • HEXIM1 exerts its inhibitory function on P-TEFb only when associated with the 7SK snRNA, while neither 7SK snRNA nor HEXIM1 alone instigates any effects. It is hypothesized that association of the 7SK snRNA with HEXIM1 leads to a conformational change that renders the cyclin T1-binding domain of HEXIM1 accessible for P-TEFb binding.
  • HEXIM1 contains several functional domains and the domains required for 7SK snRNA and cyclin T1 binding have been identified. The N-terminus of HEXIM1, AA 1-150, has been characterized as a self-inhibitory domain (ID).
  • the BR contains the binding motif for 7SK snRNA, KHRR (AAs 152-155).
  • the P-TEFb binding motif, PYNT (AAs 202-205) is located between the BR and acidic region (AR, AAs 210-250). In the absence of 7SK snRNA, the AR can interact with the adjacent BR.
  • HEXIM1 can form a homodimer or a heterodimer with a HEXIM1-related protein, HEXIM2, through the dimerization domain (DD) at the C-terminus of HEXIM1.
  • HEXIM1 stabilizes p53 by blocking the HDM2-mediated ubiquitination of p53 and plays an essential role in p53 activation induced by anti-cancer drugs/compounds.
  • HDM2 a p53-specific E3 ubiquitin ligase
  • Ubiquitination of p53 by HDM2 results in proteasome-mediated degradation of p53 protein.
  • Six lysine residues, Lys-370, -372, -373, -381, -382, and -386, located within the C-terminal domain of p53 are known as the target residues for HDM2 ubiquitination.
  • lysine residues located within the BR of HEXIM1 are identified as the major sites for HDM2 ubiquitination. Sequence alignment of the ubiquitination sites between p53 (AAs 370-386) and HEXIM1 (AAs 150-161) exhibits similar distribution of the lysine residues ( FIG. 1A ). This suggests that HEXIM1 peptide containing these ubiquitination sites may have an impact on p53 activation, resulting in p53-dependent cell arrest or cell death.
  • FGF-p53 containing the p53 ubiquitination sites (AAs 361-382) fused with a cell membrane-translocating peptide derived from Kaposi fibroblast growth factor, was reported to induce apoptosis in mutant and wild-type p53-bearing human cell lines.
  • the internalized p53 peptide is likely to compete with the endogenous p53 protein in binding to HDM2 and protect the endogenous p53 from HDM2-mediated ubiquitination, resulting in stabilization and activation of p53.
  • a FGF-BR fusion peptide was generated in the present disclosure, in which the cell penetrating FGF peptide was fused with HEXIM1 BR peptide containing the HDM2 ubiquitination residues (AAs 150-161).
  • FGF-p53 exhibited anti-proliferation effects on AML2 cells at high dosage (30 ⁇ M) but had no effects on AML3 cells ( FIG. 11 ).
  • the p53 level in AML2 has been shown to be significantly higher than that in AML3.
  • FGF-BR peptide exhibited similar cytotoxicity on both cell lines and much stronger inhibition than FGF-p53 peptide, raising the possibility that the killing mediated by FGF-BR might not depend on p53 ( FIG. 11 ).
  • Cytotoxicity was also observed in human cervical cancer cells, HeLa cells, when treated with FGF-BR peptide ( FIG. 1C ). Comparable cytotoxicity was also observed in normal cells, including HEK293 cells and human foreskin fibroblasts (HFFs) ( FIG. 1C ), suggesting the cytotoxic effect of BR peptide occurs to all cell types once it is internalized into cells.
  • HFFs human foreskin fibroblasts
  • FGF-BR peptide was also tested in human colon cancer cell line, HCT116 (p53 KO).
  • the HCT116 (p53 WT) was used as a control.
  • a fusion peptide, FGF-BR-RRR12 was generated, in which all six HDM2 ubiquitinating lysine residues were mutated to arginine and could no longer be ubiquitinated by HDM2 ( FIG. 1B ).
  • the BR-RRR12 mutant should lose its cytotoxic effects. FGF-BR-RRR12 did not exhibit reduced inhibitory effects but demonstrated stronger cell toxicity when compared to the wild-type FGF-BR peptide ( FIG. 1D ). It was also noticed that the p53 status had no effects on the cell killing mediated by FGF-BR-RRR12 ( FIG. 1D ), confirming that the cell death caused by the BR peptide was not p53-dependent.
  • Example 2 HEXIM1 BR Induced Cell Death is Independent of Apoptosis
  • the mechanism of cell death induced by the HEXIM1 BR peptide was examined.
  • the effect of FGF-BR on HCT116 (p 53 WT) and HCT116 (p 53 KO) cells was examined in a live cell imaging setting.
  • Cells treated with FGF peptide were included as a control.
  • FGF-BR peptide rapidly induced drastic changes to the cell morphology with rupturing of the plasma membrane accompanied with damages to the nuclear membrane and abnormalities to the nucleolus in both cell types ( FIG. 2A ). No effects were detected when cells were treated with FGF peptide ( FIG. 2A ).
  • Example 3 HEXIM1 BR Induces Rapid Depolarization of Mitochondrial Membrane Potential in a p53-Dependent Manner
  • a cytotoxic basic peptide, KLA elicits its killing action by disrupting the mitochondrial membrane potential (MMP), which is important for ATP generation and induction of apoptosis.
  • MMP mitochondrial membrane potential
  • HEXIM1 BR contains many basic residues, it is possible that the BR may utilize similar mechanism for its potent killing.
  • MMP was measured using JC-1 dye after incubating with FGF-X13 or FGF-BR peptides for three minutes.
  • Carbonylcyanide m-chlorophenylhydrazone (CCCP) a known mitochondrial membrane depolarizer, was used as a positive control.
  • FGF-BR treated HCT116 (p53 WT) cells experienced rapid mitochondrial depolarization within three minutes ( FIG.
  • FIG. 3A is similar to the time frame that morphological changes occurred upon addition of the FGF-BR peptide ( FIG. 2A ).
  • FIG. 3B no mitochondrial depolarization was observed in HCT116 (p 53 KO) cells treated with FGF-BR ( FIG. 3B ), although these cells encountered rapid cytotoxicity similar to HCT116 (p53 WT) cells ( FIG. 2A ). It was known that p53 would translocate to the mitochondria, resulting in the reduction of MMP in p53-mediated apoptosis.
  • HEXIM1 BR peptide is likely to induce cell killing though an alternative pathway that acts independently of p53 and apoptosis.
  • NPM is a multi-functional protein and participates in the process of ribosome biogenesis. In addition to its role in protein translation, NPM is required to maintain DNA integrity in cells. Knockout of NPM results in accumulation of DNA damage, which clearly indicates the essential role of NPM in cell proliferation and survival.
  • NPM is located in nucleolus, the sub-cellular site of ribosome synthesis and assembly. About 35% of AML patients carry the cytoplasmic-misallocated mutant form of NPM, NPMc+. NPMc+ is found to interact and sequester a portion of HEXIM1 in the cytoplasm of the NPMc+ AML cell line and activates P-TEFb-dependent transcription, suggesting the involvement of HEXIM1 in tumorigenesis of AML.
  • the BR domain of HEXIM1 was known to contain a nucleolar localization sequence.
  • the BR was fused with yellow fluorescent protein (YFP)
  • YFP yellow fluorescent protein
  • the BR-YFP was found to localize in nucleoli.
  • NPM was identified as a HEXIM1 binding protein, and the BR of HEXIM1 was required for NPM binding.
  • immunofluorescence was performed to examine the sub-cellular distribution of NPM in the FGF-BR-treated HCT116 (p53 WT) and HCT116 (p53 KO) cells. Normal nucleolar localization of NPM was observed in control experiments ( FIG.
  • Therapeutic peptides can be divided into two major categories: cell targeting and cytotoxic peptides.
  • Targeting peptides including the cancer targeting peptides, provide selectivity and specificity to direct the targeted treatment.
  • Cytotoxic peptides can be used as the toxin/payload in fusion peptides, nano-particle conjugates, and antibody drug conjugates (ADCs) for potential cancer treatment.
  • ADCs antibody drug conjugates
  • a breast cancer targeting peptide LTV
  • LTV-BR and LTV-BR-RRR12 SEQ ID NO: 16
  • Both LTV-BR and LTV-BR-RRR12 peptides exhibited anti-proliferation effects in breast cancer cell lines MCF7 and MDA-MB-231, while no effects were detected using the LVT-X13 control peptide ( FIGS. 5A and 5B ).
  • MDA-MB-231 cells are triple-negative with the absence of expression of oestrogen receptor, progesterone receptor and HER2, LTV-BR and LTV-BR-RRR12 elicited similar anti-cancer activity as compared to MCF7 cells ( FIG. 5B ).
  • untagged control X13 SEQ ID NO: 12
  • the two BR peptides had no effect on cell viability ( FIGS. 5C and 5D ).
  • KLA peptide (SEQ ID NO: 13), a cytotoxic peptide, was often fused with a cancer-targeting peptide or conjugated to antibodies recognizing cancer cells (as antibody-drug conjugate, ADC) to exert cell-killing effect in anti-cancer therapy.
  • LTV-KLA peptide (SEQ ID NO: 17) was generated and its effect was compared to that of LTV-BR.
  • LTV-KLA exhibited stronger inhibition on the viability of MDA-MB-231 cells ( FIG. 5B ).
  • untagged KLA also showed non-specific killing on MDA-MB-231 cells, while BR- or BR-RRR12-treated cells remained highly viable ( FIG. 5D ).
  • LTV-KLA also exhibited non-specific cell killing on non-breast cancer cell lines such as CHO (Chinese hamster ovary) and OPM-2 (multiple myeloma) cells, while little or no effects were observed in the LTV-BR-treated cells ( FIGS. 9A and 9B ). Similar observations were seen for normal human fibroblasts HFF and WI-38, indicating the specificity of LTV-BR towards breast cancer cell lines but not LTV-KLA ( FIGS. 9C and 9D ). It is predicted that a portion of the KLA ADCs will be broken down before reaching the target cancer cells, even though non-cleavable linkers may be used to generate these conjugates.
  • KLA peptides may be released from the conjugates, resulting in an off-target killing of normal cells/tissues.
  • the use of KLA as the toxic load in cancer therapy is questionable due to its non-specificity.
  • untagged HEXIM1 BR peptide is unable to kill cells when it is not fused with any cell penetrating or targeting peptides, suggesting that HEXIM1 BR peptide may be a safer alternative as compared to KLA, for the development of anti-cancer therapeutics.
  • the HEXIM1 BR peptide did not cause any cytotoxic effects when it was not fused with cell penetrating (for example, FGF) or cancer cell targeting (for example, LTV) peptides. It is thus possible that the unfused BR peptide may fail to internalize into cells without specific guidance. To test this hypothesis, fluorescent-labelled BR and LTV-BR peptides were generated and their presence in cells was examined by confocal microscope.
  • MCF7 cells were incubated with fluorescent-labeled peptides, followed by a washing step to remove the peptides failing to penetrate into cells.
  • No fluorescent signals were observed in HEXIM1 BR peptide-treated cells ( FIG. 6A ), indicating that the BR peptide was not internalized.
  • fluorescent signals were detected within the cells ( FIG. 6 ).
  • LTV-BR was readily internalized into MCF7 cells and distributed in cytoplasm and nuclei ( FIG. 6A ). It was noted that its strong fluorescent signals were primarily localized in the nucleoli ( FIG. 6A , LTV-BR-FITC).
  • Detection of fluorescent signals in KLA-treated cells helps to explain the non-specific cytotoxicity induced by KLA peptide ( FIG. 6A ), while no fluorescent signal was observed in HEXIM1 BR-treated cells, indicating that the BR peptide could not enter the cells by itself ( FIG. 6A , BR-FITC).
  • Cells treated with LTV-KLA demonstrated that the sub-cellular localization of the peptide was observed mainly in the cytoplasm ( FIG. 6A ).
  • the different distribution of LTV-BR and LTV-KLA suggests that BR and KLA may utilize different mechanisms for cell killing.
  • LTV peptide directed the uptake of almost 100% of LTV-fused peptides (LTV-BR and LTV-KLA) into the breast cancer cell line ( FIG. 6B ).
  • LTV-BR and LTV-KLA LTV-fused peptides
  • FIG. 6B Approximately 65% of KLA-FITC-treated MCF7 cells internalized KLA-FITC, whereas there was no entry of BR-FITC into MCF7 cells ( FIG. 6B ).
  • mice fibroblast cells 3T3 2.5 ⁇ 10 4 cells/ml of mouse fibroblast cells 3T3 were cultured in gelatin coated plates in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (Gibco) to reach 100% confluent. The cells were then maintained at 100% confluency by supplementing with fresh media in a 37° C. incubator with 5% CO 2 for 3 days.
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS fetal bovine serum
  • Gibco penicillin/streptomycin
  • DMI Induction Media which is DMEM supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin, 0.1% 3-isobutyl-1-methylxanthine (sigma), 0.1% insulin (sigma) and 0.1% dexamethasone (sigma)
  • Insulin Induction Media which is DMEM supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin and 0.1% insulin.
  • the insulin induction media was changed every 2 days until the cells were differentiated to adipose cells.
  • a D-form BR peptide with the sequence of QLGKKKHRRRPSKKKRHW was tagged to a fat tissue targeting peptide of the sequence CKGGRAKDC (ATS peptide, SEQ ID NO: 39) to generate the ATS-BR D peptide of the sequence CKGGRAKDCGG(QLGKKKHRRRPSKKKRHW) D (SEQ ID NO: 40). Both undifferentiated 3T3 cells and differentiated adipose cells were treated with the ATS-BR D peptide. ATS-tagged KLA D peptide was used as a positive control. Cell viability results shown in FIG.
  • ATS-BR D peptide has no cytotoxic effect on undifferentiated 3T3 mouse fibroblast cells, but has significant cytotoxic effect on differentiated adipose cells.
  • the cytotoxic effect of ATS-BR D peptide on adipose cells is of similar potency as ATS-KLA D peptide.
  • Example 8 Use of HEXIM1 BR Peptide as the Toxin for Antibody Drug Conjugates (ADCs)
  • the cytotoxic HEXIM1 BR peptides are covalently attached to antibodies through chemical linkers.
  • Cysteine and lysine are two most naturally occurring amino acids which are used to attach the toxin through the linker to the antibody.
  • SPDP N-succinidyl-3-(2-pyridylothio)propionate
  • SCC succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate
  • a cysteine residue is added at the N- or C-terminal of the BR peptide. After removing excess linkers and peptides, the effects of the BR-conjugated ADCs are analyzed in vitro using the selected breast cancer cell lines.
  • IgGs have four pairs of interchain disulfide bonds, two between the heavy chains in the hinge region and two on Fab between heavy and light chains. Intrachain disulfide bonds are to be left intact because they are critical to maintain the basic IgG domain structure essential for antigen recognition. Only partial reduction of the interchain disulfide bonds at the hinges gives eight potential conjugation sites through cysteine residues.
  • Human cell lines including HeLa, HEK293, MCF7, MDA-MB-231, CHO-K1, OPM-2, and WI-38 were obtained from American Type Culture Collection.
  • AML2 and AML3 cells were purchased from Deutsche Sammlung von Mikroorganismen und Zellkulturen.
  • HCT116 p53+/+ and p53 ⁇ / ⁇ cells were kindly given by Dr. Bert Vogelstein.
  • Primary human foreskin fibroblasts (HFF) were obtained from Dr. Mark Stinski.
  • HCT116, HCT116 (p53 KO), HeLa, 293, HFF, MCF7, CHO-K1, and WI-38 cells were cultured in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (Gibco).
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS fetal bovine serum
  • penicillin/streptomycin Gibco
  • MDA-MB-231 and OPM-2 cells were cultured in RPMI-1640 (Gibco) containing 10% FBS and 1% penicillin/streptomycin. All cells were routinely maintained in a 37° C. incubator with 5% CO 2 .
  • Cells were lysed in lysis buffer [50 mM Tris-HCl, (pH 7.5), 150 mM NaCl, 1% NP40, 0.5% sodium deoxycholate, Protease Inhibitor tablet (Roche)] and used for western blotting.
  • Western blotting was carried out as previously mentioned.
  • the primary antibodies used include anti-NPM (Invitrogen) and anti-actin (Millipore).
  • the film of western blot was scanned, and the protein bands were quantified by the GS-800 densitometer (Bio-Rad). The protein level of NPM was quantified after normalizing with the loading control, actin.
  • Cells were plated in clear-bottomed white walled 96-well plates (Corning) and incubated overnight. Cells were treated with indicated peptides in 1% FBS-containing media for overnight or indicated timings at 37° C. Cell viability was measured with CellTiter-Glo reagent (Promega) according to the manufacturer's instructions. For cytotoxicity assay, cells were plated in black walled 96-well plates (Corning) to allow them to adhere overnight.
  • cytotoxicity induced was determined by CellTox cytotoxicity assay (Promega) 30 minutes after addition of peptides according to the manufacturer's instructions. Luminescence was determined using an Infinite 200 multiplate reader (Tecan).
  • JC-1 1,1′,3,3′-tetraethylbenzamidazolocarbocyanin iodide
  • JC-1 was dissolved in DMSO (200 ⁇ M).
  • Peptide-treated cells in 96-well deep sided, clear bottom, dark sided microplates were incubated with media containing JC-1 for 30 min at 37° C. and then washed twice with warm PBS. Changes in MMP were determined using a multiplate spectrofluorometer (Tecan) (excitation: 475 nm; emission: 530 nm (green); emission: 590 nm (red)). The decrease in the ratio of red to green fluorescence was used to determine relative mitochondrial depolarization.
  • MCF7 cells were plated on 6-cm culture dish to allow overnight adherence.
  • FITC-labeled peptides (30 ⁇ M) were added to the cells, incubated for 30 min at 37° C., and subsequently washed three times with PBS. The cells were then trypsinized, collected by centrifugation, and finally resuspended in 500 ⁇ l ice-cold 2% FBS-containing PBS for flow cytometry analysis.
  • MCF7 cells were plated on 4-chamber glass cover slides (Lab-Tek) to adhere overnight, incubated with FITC-labeled peptides (30 ⁇ M) for 30 min, and then washed three times with PBS before being fixed and mounted with DAPI-containing mounting solution (Vectashield). Images were acquired using a Nikon AIR confocal laser scanning microscope equipped with a 60 ⁇ oil-immersion objective lens (SBIC-Nikon Imaging Centre).

Abstract

Disclosed are isolated cytotoxic peptides having similar sequences as the basic region (BR) of Hexamethylene Bisacetamide Inducible Protein 1 (HEXIM1). Preferred embodiments include QLGKKKHRRRPSKKKRHW (SEQ ID No: 3). QLGRRRHRRRPSRRRRHW (SEQ ID No: 4) and QLGKKILAARPSKKKRHW (SEQ ID No: 5). Also encompassed are isolated nucleic acid molecules encoding for the claimed peptides, vectors comprising the isolated nucleic acids, compositions comprising peptides conjugated to cell-targeting or penetrating peptides or antibodies, nucleic acid molecules or vectors expressing conjugates thereof; methods of treating or preventing diseases or conditions, including cancers and obesity as well as a method of eliminating undifferentiated stem cells.

Description

CROSS-REFERENCE TO RELATED APPLICATIONS
This patent application is a U.S. National Phase Application under 35 U.S.C. § 371 of International Application No. PCT/SG2016/050101, filed on 3 Mar. 2016, entitled CYTOTOXIC HEXIM1 PEPTIDES AND USES THEREOF, which claims priority to Singapore provisional application Nos. 10201501642V filed on 4 Mar. 2015 and 10201504803T filed on 17 Jun. 2015, the contents of which were incorporated by reference in their entirety for all purposes.
INCORPORATION BY REFERENCE
This application incorporates by reference the material (i.e., Sequence Listing) in the ASCII text file named 9869SG3622 corrected ST25_3868247_1.txt, created on Jan. 4, 2018, having a file size of 172,132 bytes.
FIELD OF THE INVENTION
The present invention generally relates to the field of biochemistry. In particular, the present invention relates to cytotoxic peptides, compositions containing cytotoxic peptides and the use of the peptides and compositions in the prevention and treatment of cancers.
BACKGROUND OF THE INVENTION
Cancer is the second leading cause of death worldwide, accounting for 8.2 million deaths in 2012. It is expected that annual cancer cases will rise from 14 million in 2012 to 22 million within the next two decades.
Conventional treatments for cancer using radiation or chemotherapeutic drugs target rapidly proliferating cells and induce such cells to undergo apoptosis. Most chemotherapeutic agents exhibit some degree of toxicity toward normal cells at therapeutic doses, causing undesired side effects that may be dose limiting, thereby reducing the usefulness of the drugs. Furthermore, these traditional methods of treatment are not successful in treating many types of cancers, particularly those that are resistant to apoptotic stimuli. Since the cytotoxic agents in most chemotherapeutic drugs act primarily through p53-dependent induction of apoptosis, and p53 is often mutated in cancers cells, resistance to chemotherapy is often observed in cancer patients. As a consequence, there is an ongoing need for new therapeutic agents that are more effective in treating cancers.
SUMMARY OF THE INVENTION
In one aspect, the present invention refers to an isolated cytotoxic peptide having sequence identity of at least 66% to SEQ ID NO: 3 or functional part thereof.
In another aspect, the present invention refers to an isolated cytotoxic peptide comprising at least one amino acid sequence represented by the consensus sequence Q1-L2-G3-X4-X5-X6-X7-X8-X9-R10-P11-S12-X13-X14-X15-R16-H17-W18 (SEQ ID NO: 36), or functional part thereof, wherein X is any amino acid.
In one aspect, the present invention refers to an isolated nucleic acid molecule comprising the nucleotide sequence encoding the isolated cytotoxic peptide as described herein.
In another aspect, the present invention refers to a vector comprising the isolated nucleic acid molecule as described herein.
In another aspect, the present invention refers to a pharmaceutical composition comprising the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, or the vector as described herein.
In yet another aspect, the present invention refers to a method of treating or preventing cancer in a subject, comprising administering the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, the vector or the pharmaceutical composition as described herein.
In one aspect, the present invention refers to a method of eliminating undifferentiated stem cells in a subject having stem cell transplantation, comprising administering the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, the vector or the pharmaceutical composition as described herein.
In another aspect, the present invention refers to a method of treating or preventing obesity in a subject, comprising administering the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, the vector or the pharmaceutical composition as described herein.
BRIEF DESCRIPTION OF THE DRAWINGS
The invention will be better understood with reference to the detailed description when considered in conjunction with the non-limiting examples and the accompanying drawings, in which:
FIG. 1 shows FGF-tagged BR peptide (SEQ ID NO: 19) and FGF-tagged mutant BR peptides induce cell death in various cell lines. FGF tag is a cell membrane-translocating peptide derived from Kaposi fibroblast growth factor. (A) Alignment of the HDM2 ubiquitination sites of p53 (amino acids (AAs) 370-386, SEQ ID NO: 34) and HEXIM1 (AAs 150-161, SEQ ID NO: 35) shows that ubiquitin sites of p53 and HEXIM1 exhibit similar distribution of the lysine residues (the HDM2-ubiquitinated lysine residues are underlined). (B) Alignment of wild-type and mutant HEXIM1 BR peptides (RRR12 and ILAA) indicates the amino acid residues that have been mutated (the mutated residues are underlined). (C) Cell viability plots showing the effects of FGF-tagged BR peptide in various cancer cell lines. Cancer cell lines: Acute Myeloid Leukemia cell lines—AML2, AML3, cervical cancer cell line—HeLa, and non-cancer cell lines—Human Embryonic Kidney 293 (293) and Human Foreskin Fibroblast (HFF) cell lines were treated overnight with various concentrations of indicated FGF-fused peptides before cell viability was determined by Cell-Titer Glo assays. Cells treated with the FGF-X13 peptide (SEQ ID NO: 18) were used as a negative control. The results show that FGF-tagged BR peptide induces cell death in AML2, AML3, HeLa, HEK293 and HFF cell lines. (D) Cell viability plots showing the effects of FGF-tagged BR peptide and variants thereof in human colon cancer cell line HCT116 with wild-type p53 (p53 WT) or p53 knockout (p53KO). HCT116 (p53 WT) and HCT116 (p53 KO) cells were treated with FGF-X13, FGF-BR, FGF-BR-RRR12 (SEQ ID NO: 20) and FGF-BR-ILAA (SEQ ID NO: 21) peptides at various concentrations overnight before cell viability assays were performed. Data representative of at least three independent experiments performed in triplicate were shown with values expressed as mean±SD. The results indicate that FGF-tagged BR peptide shows similar level of cytotoxicity in HCT116 p53 wild-type (p53 WT) and HCT116 p53 knockout (p53 KO) cell lines. FGF-tagged mutant BR peptides, BR(RRR12) and BR(ILAA) also show cytotoxicity in both HCT116 (p53 WT) and HCT116 (p53KO) cell lines.
FIG. 2 shows FGF-/LTV-tagged BR peptide induced rapid cell death independently of apoptosis. (A) shows representative time-lapse differential interference contrast (DIC) snapshots of treated HCT116 (p53 WT) and HCT116 (p53 KO) cells (bar represents 100 μm). HCT116 (p53 WT) and HCT116 (p53 KO) cells were cultured on glass slides overnight, treated with vehicle control (0.5% dimethyl sulfoxide (DMSO)) or the indicated FGF peptides (30 μM). Time-lapse DIC imaging highlights dynamic morphological changes in treated HCT116 cells by spinning disk confocal microscopy. The results indicate that in both HCT116 (p53 WT) and HCT116 (p53KO) cell lines, FGF tag alone does not affect morphology or viability of the cells. In contrast, FGF-BR peptide rapidly induces drastic changes to the cell morphology with rupturing of the plasma membrane accompanied with damages to the nuclear membrane and abnormalities to the nucleolus in both cell types. (B) cytotoxicity fluorescence plots of breast cancer cells MCF7 showing the effect of pan-caspase inhibitor z-VAD-Fmk on cells treated with LTV-tagged BR peptide. LTV is a breast cancer cell targeting peptide. MCF7 cells were incubated with or without z-VAD-Fmk (for 90 min) and then with indicated LTV-tagged peptides (30 μM) for thirty minutes at 37° C. Cells treated with LTV-X13 peptide (SEQ ID NO: 14) or vehicles, DMSO (0.5%), were used as negative controls. Treated cells were subjected to the cytotoxicity assay as described in Example 9—Materials and Methods. Data representative of at least three independent experiments performed in triplicate were shown with values expressed as mean±SD. The results indicate that LTV-tagged BR (SEQ ID NO: 15) has significant cytotoxic effect on MCF7 cell line, and the effect could not be inhibited by z-VAD-Fmk.
FIG. 3 shows FGF-tagged BR peptide decreases mitochondrial membrane potential in HCT116 (p53 WT) cell line but not in HCT116 (p53 KO) cell line. (A) shows bar chart of mitochondrial membrane potential in HCT116 (p53 WT) cell line treated with negative and positive controls and the FGF-tagged BR peptide. (B) shows bar chart of mitochondrial membrane potential in HCT116 (p53 KO) cell line treated with negative and positive controls and the FGF-tagged BR peptide. HCT116 (p53 WT) and HCT116 (p53 KO) cells were cultured in 96-well plates, treated with indicated FGF-tagged peptides (30 μM) for three minutes. A mitochondrial membrane depolarizer, carbonyl cyanide 3-chlorophenylhydrazone (CCCP), was used as a positive control. Cells treated with FGF-X13 peptide or vehicle, DMSO (0.5%), was used as negative controls. Treated cells were subjected to mitochondrial membrane potential (MMP) measurement using JC-1 fluorescence-based assay for six independent experiments. Results were summarized as mean±SD (***, P<0.0001; ns: not significant, Student's t test). The results indicate that FGF-tagged BR peptide decreased mitochondrial membrane potential in HCT116 cell line is dependent on p53.
FIG. 4 shows FGF-tagged BR peptide alters the sub-cellular localization and protein level of endogenous NPM. (A) representative immunofluorescence images showing the sub-cellular localization of α-NPM (bar represents 10 μm). HCT116 (p53 WT) and HCT116 (p53KO) cells were cultured on glass slides overnight, treated with FGF-X13 or FGF-BR (30 μM). Cells treated with FGF-X13 peptide or vehicle, DMSO (0.5%), was used as controls. Treated cells were immunostained with an anti-NPM antibody and analyzed by laser scanning confocal microscopy (Zeiss). Nuclei were visualized by staining with 4′,6-diamidino-2-phenylindole (DAPI). The results indicate that NPM is mislocated into the cytoplasm in cells treated FGF-tagged BR peptide, in both HCT116 (p53 WT) and HCT116 (p53 KO) cell lines. (B) results of western blot showing the expression level of α-NPM protein (α-actin was used as the internal control. HCT116 cells were plated overnight prior to addition of vehicle control (0.5% DMSO) or indicated peptides (30 μM) at 37° C. Lysates were subsequently harvested and subjected to western blotting with anti-NPM and anti-actin antibodies. The level of NPM protein was quantified as described in Example 9—Materials and Methods. The results show that endogenous level of NPM is reduced in cells treated with FGF-tagged BR peptide, in both HCT116 (p53 WT) and HCT116 (p53 KO) cell lines.
FIG. 5 Cell viability plots showing the effects of LTV-tagged BR peptide and variants thereof, as well as untagged BR peptide and variants thereof, in two breast cancer cell lines, (A) MCF7 and (B) MDA-MB-231. Cells were treated with indicated LTV-tagged peptides at different concentrations overnight before cell viability assays were performed. Both LTV-tagged BR and LTV-tagged BR (RRR12) peptides decreased cell viability in (A) MCF7 and (B) MDA-MB-231 cell lines. Effects of the indicated un-tagged peptides on the viability of (C) MCF7 and (D) MDA-MB-231 cells were analyzed by cell viability assays. The untagged X13 was used as a negative control. The results indicate that untagged BR and BR (RRR12) peptides have no effect on cell viability in (C) MCF7 and (D) MDA-MB-231 cell lines. Data representative of at least three independent experiments performed in triplicate were shown with values expressed as mean±SD. The results indicate that LTV-tagged BR peptide decreases cell viability in breast cancer cells, while BR peptide alone shows no effect on cell viability.
FIG. 6 shows the untagged HEXIM1 BR peptide fails to enter cells by itself, while another cytotoxic peptide, KLA, can enter the cell without a cell penetrating and/or cell targeting tag. (A) shows images of confocal microscopy of MCF7 cells treated with vehicle control and the indicated peptides (bar represents 10 μm). MCF7 cells were cultured on glass slides overnight before incubation with the indicated FITC-labeled peptides (30 μM) for 30 min and subsequently analyzed by laser scanning confocal microscopy. Nuclei were visualized by DAPI. Cells treated with vehicle, DMSO (0.5%) were used as control. Pointed arrow indicates FITC fluorescence. The results show that no FITC fluorescence can be defected in cells treated with BR-FITC peptide, while FITC fluorescence can be detected in cells treated with LTC-BR-FITC, KLA-FITC and LTV-KLA-FITC peptides. (B) shows bar chart of FITC-positive cells quantified by flow cytometry. Data represent percentage of fluorescence-positive cells in total cell population. Results were summarized as mean±SD of three independent experiments (***, P<0.0001, Student's t test). The results indicate that no significant cellular uptake of BR-FITC was observed, while the cellular uptake of KLA-FITC, LTV-BR-FITC and LTV-KLA-FITC is significantly higher compared to BR-FITC.
FIG. 7 shows cell viability plots (upper panel) of cells treated control, LTV-tagged BR and mutated variants thereof (sequences indicated in the lower panel). MDA-MB-231 breast cancer cells were treated with indicated truncated LTV-tagged peptides at various concentrations overnight before cell viability assays were performed. Cells treated with LTV-X13 were used as negative control, cells treated with LTV-BR were used as positive control. Data representative of at least three independent experiments performed in triplicate were shown with values expressed as mean±SD. The results indicate that LTV-tagged BR peptide requires all three stretches of basic residues (KKK, RRR and KKK) to establish full potency in cell killing.
FIG. 8 shows cell viability plots (upper panel) of cells treated control, LTV-tagged BR and alanine substituted variants thereof (sequences indicated in the lower panel). MDA-MB-231 breast cancer cells were treated with indicated alanine-substituted LTV-tagged peptides at 10 μM and 30 μM overnight before cell viability assays were performed. Cells treated with LTV-X13 were used as negative control, cells treated with LTV-BR were used as positive control. Data representative of at least three independent experiments performed in triplicate were shown with values expressed as mean±SD. The results indicate that alanine residue substitution in the middle and C-terminal regions of the HEXIM1 BR peptide abolished the cytotoxic activity of LTV-BR.
FIG. 9 shows cell viability plots of various cell lines treated with control and LTV-tagged BR and LTV-tagged KLA peptides. Chinese hamster ovary cancer cell line, CHO-K1; multiple myeloma cell line, OPM-2, human foreskin fibroblasts, HFF; and human lung fibroblasts, WI-38 were treated with indicated LTV-fused peptides at various concentrations overnight before cell viability assays were performed. Cells treated with LTV-X13 was used as a negative control. Data representative of at least three independent experiments performed in triplicate were shown with values expressed as mean±SD. The results indicate that LTV-tagged KLA peptide exhibits non-specific killing on non-breast cancer cell lines and normal human fibroblasts, while LTV-tagged BR peptide does not show such non-specific cytotoxicity.
FIG. 10 shows cell viability plots of (A) undifferentiated 3T3 cells and (B) adipose cells treated with BRD and KLAD peptides tagged with lipocyte-targeting peptide, ATS (ATS-BRD and ATS-KLAD peptides). The results indicate that the ATS-BRD peptide has no cytotoxic effect on undifferentiated 3T3 mouse fibroblast cells, but has significant cytotoxic effect on differentiated adipose cells. The cytotoxic effect of ATS-BRD peptide on adipose cells is of similar potency as ATS-KLAD peptide.
FIG. 11 shows that the cytotoxic effect exerted by BR is independent of p53. AML2 and AML3 cells were treated with FGF-tagged p53 and FGF-tagged BR peptides at the indicated concentrations. Cell viability was measured 16 hours post-incubation by Cell-Titer Glo. Cells treated with vehicle, DMSO (0.5%), were used as control. (A) Cell viability plots showing the effects of FGF-tagged p53 and FGF-tagged BR peptides in AML2 cell line. (B) Cell viability plots showing the effects of FGF-tagged p53 and FGF-tagged BR peptides in AML3 cell line.
DETAILED DESCRIPTION OF THE PRESENT INVENTION
In the search for new therapeutic agents that are more effective in treating cancers, it was found that the basic region (BR) of hexamethylene bisacetamide inducible protein 1 (HEXIM1, represented by SEQ ID NO: 1) exerts its cytotoxic effects independent of the status of p53. Thus, the present disclosure provides an isolated peptide that has the cytotoxic activity of the BR of HEXIM1.
In one example, there is provided an isolated cytotoxic peptide having sequence identity of at least 66% to SEQ ID NO: 3, or functional part thereof. In some examples, the isolated cytotoxic peptide has sequence identity of at least 72%, at least 77%, at least 83%, at least 88% or at least 94% to SEQ ID NO: 3, or functional part thereof. In some other examples, the isolated cytotoxic peptide has sequence identity of 66% to 78%, 77% to 89%, 88% to 95% to SEQ ID NO: 3, or functional part thereof.
As used herein, the basic region (BR) of HEXIM refers to the region between AAs 150-180 of HEXIM1, which includes a stretch of basic residues. The BR is represented by the sequence of KKKHRRRPSKKKRHWKPYYKLTWEEKKKFDE (SEQ ID NO: 2).
As used herein, an “isolated” peptide is intended to mean a peptide removed from its native environment, such as recombinantly produced peptides expressed in host cells and native or recombinant peptides which have been substantially purified by any suitable technique. Isolated peptides according to the present disclosure further include such compounds produced synthetically.
The terms “peptide” used herein refers to polymers of amino acid residues of any length. The polymer may be linear or branched, it may comprise modified amino acids or amino acid analogues, and it may be interrupted by chemical moieties other than amino acids. The terms also encompass an amino acid polymer that has been modified naturally or by intervention, for example disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labelling or bioactive component. The term peptide encompasses two or more naturally occurring or synthetic amino acids linked by a covalent bond (for example, an amide bond). The peptide as disclosed herein can be of various lengths, such as having 6 to 200, 8 to 190, 10 to 180, 12 to 170, 14 to 160, 16 to 150, 18 to 140, 20 to 130, 22 to 120, 24 to 110, 26 to 100, 28 to 90, 30 to 80, 32 to 70, 34 to 60, 36 to 50 or 38 to 45 amino acid residues.
As used herein, the term “amino acid” is defined as having at least one primary, secondary, tertiary or quaternary amino group, and at least one acid group, wherein the acid group may be a carboxylic, sulfonic, or phosphoric acid, or mixtures thereof. The amino groups may be “alpha”, “beta”, “gamma” to “omega” with respect to the acid group(s). Suitable amino acids include, without limitation, both the D- and L-isomers of the 20 common naturally occurring amino acids found in peptides (for example, A, R, N, C, D, Q, E, G, H, I, L, K, M, F, P, S, T, W, Y, V (as known by the one-letter or three letter abbreviations)) as well as the naturally occurring and unnaturally occurring amino acids prepared by organic synthesis or other metabolic routes.
The term “sequence identity” as used herein refers to a relationship between two or more peptide sequences, namely a reference sequence and a given sequence to be compared with the reference sequence. Sequence identity is determined by comparing the given sequence to the reference sequence after the sequences have been optimally aligned to produce the highest degree of sequence similarity, as determined by the match between strings of such sequences. Upon such alignment, sequence identity is ascertained on a position-by-position basis, for example, the sequences are “identical” at a particular position if at that position, the amino acid residues are identical. The total number of such position identities is then divided by the total number of amino acid residues in the reference sequence to give % sequence identity. Sequence identity can be readily calculated by known methods. Some methods used to determine the sequence identity are designed to give the largest match between the sequences tested. Methods to determine sequence identity are codified in publicly available computer programs that determine sequence identity between given sequences. These programs optimally align sequences using default gap weights in order to produce the highest level of sequence identity between the given and reference sequences. As an illustration, by a peptide or polypeptide having a given amino acid sequence having at least, for example, 66% sequence identity to a reference amino acid sequence, it is intended that the given amino acid sequence of the peptide or polypeptide is identical to the reference sequence except that the given peptide or polypeptide sequence may include up to 34 amino acid alterations per each 100 amino acids of the reference amino acid sequence. In other words, to obtain a given peptide or polypeptide sequence having at least 66% sequence identity with a reference amino acid sequence, up to 34% of the amino acid residues in the reference sequence may be deleted or substituted with another amino acid, or a number of amino acids up to 34% of the total number of amino acid residues in the reference sequence may be inserted into the reference sequence. These alterations of the reference sequence may occur at the amino or the carboxyl terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in the one or more contiguous groups within the reference sequence. In some examples, residue positions that are not identical differ by conservative amino acid substitutions. However, conservative substitutions are not included as a match when determining sequence identity. For example, the isolated cytotoxic peptide QLGKKILAARPSKKKRHW (SEQ ID NO: 5) has 77.8% sequence identity with SEQ ID NO: 3; the isolated cytotoxic peptides QLGRRRHRRRPSKKKRHW (SEQ ID NO: 4) and HRRRPSKKKRHW (SEQ ID NO: 37) both have 66.7% sequence identity with SEQ ID NO: 3, the isolated cytotoxic peptide QLGAAAHRRRPSKKKRHW (SEQ ID NO: 38) has 83.3% sequence identity with SEQ ID NO: 3.
Thus, in some examples, the isolated cytotoxic peptide as described herein has at least 12, at least 13, at least 14, at least 15, at least 16 or at least 17 amino acid residues that are the same as SEQ ID NO: 3, or functional part thereof. In some other examples, the isolated cytotoxic peptide has 12 to 14 or 15 to 17 amino acid residues that are the same as SEQ ID NO: 3, or functional part thereof.
The terms “cytotoxic” and “cytotoxicity” as used herein refer to the effect of a compound or composition of being toxic to the cells. Effects of cytotoxicity on cells include, but are not limited to, cell swelling, cell lysis, loss of cell membrane integrity, decreased rate of metabolism, decreased cell viability, cell deaths (by inducing apoptosis or necrosis). Thus, the cytotoxic effects can be exerted via various pathways. Examples of possible pathways include, but are not limited to, apoptosis-independent pathways, positive transcription elongation factor b (P-TEFb)-independent pathway, p53-independent pathway. The terms “low toxicity” or “reduced toxicity” as used in the present disclosure refer to a decrease in toxicity towards non-cancerous cells and tissue relative or compared to cancerous cells and tissue. In some examples, the low or reduced toxicity towards non-cancerous cells and tissue is at most about 0.9 times, at most about 0.8 times, at most about 0.7 times, at most about 0.6 times, at most about 0.5 times, at most about 0.4 times, at most about 0.3 times, at most about 0.2 times, at most about 0.1 times, or at most about 0.05 times the toxicity towards cancer cells and tissue samples obtained from the same or a different subject.
The term “apoptosis” as used herein refers to programmed cell death, characterized by cell changes such as blebbing, cell shrinkage, nuclear fragmentation, chromatin condensation, chromosomal DNA fragmentation, and global mRNA decay.
As used herein, the term “functional part” of a peptide or amino acid sequence refers to the part of the peptide or the amino acid sequence that produces the overall function of a peptide or amino acid sequence. In some examples, the functional part of a peptide or amino acid sequence comprises 6 to 17, 7 to 16, 8 to 15, 9 to 14, 10 to 13 or 11 to 12 amino acid residues of the peptide or amino acid sequence. In one example, the functional part of SEQ ID NO: 3 refers to the part of SEQ ID NO: 3 that has cytotoxic effect. In some specific examples, the functional part of SEQ ID NO: 3 comprises at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, at least fifteen, at least sixteen or at least seventeen amino acid residues of the peptide or amino acid sequence. In some other examples, the functional part of SEQ ID NO: 3 comprises at least the following stretches of amino acid residues RRR (AAs 8-10 of SEQ ID NO: 3) and KKK (AAs 13-15 of SEQ ID NO: 3). Thus, some examples of the isolated cytotoxic peptide as described herein comprises amino acid residues at positions corresponding to amino acids 8 to 10 and 13 to 15 of SEQ ID NO: 3. Exemplary peptides include, but are not limited to, the sequences of:
i)
(SEQ ID NO: 3)
QLGKKKHRRRPSKKKRHW;
ii)
(SEQ ID NO: 37)
HRRRPSKKKRHW;
and
ii)
(SEQ ID NO: 38)
QLGAAAHRRRPSKKKRHW.
The isolated cytotoxic peptide as described above may comprise at least one amino acid sequence that can be represented by a consensus sequence. Thus, in one example, there is provided an isolated cytotoxic peptide having sequence identity of at least 66% to SEQ ID NO: 3, or functional part thereof, comprising at least one amino acid sequence represented by the consensus sequence Q1-L2-G3-X4-X5-X6-X7-X8-X9-R10-P11-S12-X13-X14-X15-R16-H17-W18 (SEQ ID NO: 36), or functional part thereof. In some examples, the isolated cytotoxic peptide has sequence identity of at least 72%, at least 77%, at least 83%, at least 88% or at least 94% to SEQ ID NO: 3, or functional part thereof. In some other examples, the isolated cytotoxic peptide has sequence identity of 66% to 78%, 77% to 89%, 88% to 95% to SEQ ID NO: 3, or functional part thereof.
In another example, there is provided an isolated cytotoxic peptide comprising at least one amino acid sequence represented by the consensus sequence Q1-L2-G3-X4-X5-X6-X7-X8-X9-R10-P11-S12-X13-X14-X15-R16-H17-W18 (SEQ ID NO: 36), or functional part thereof.
The term “consensus sequence” as used herein refers to an amino acid sequence determined by aligning a series of multiple sequences. It defines an idealized sequence that represents the predominant choice of amino acid at each corresponding position of the multiple sequences.
In some examples, X4, X5 and X6 can be any amino acids or may be deleted. In some examples, X4, X5 and X6 are positively charged amino acids. In some examples, X4, X5 and X6 are the same. In some other examples, at least two of X4, X5 and X6 are different from each other. In some examples, X7 can be histidine (H) or leucine (L). In some examples, X8 and X9 are positively charged amino acids. In some other examples, X8 and X9 can be small amino acid residues including but are not limited to alanine (A). In some examples, X8 and X9 are the same, while in some other examples, X8 and X9 are different. In some examples, X13, X14 and X15 are positively charged amino acids. In some examples, X13, X14 and X15 are the same. In some other examples, at least two of X13, X14 and X15 are different from each other.
In general, the positively charged amino acids may include, but are not limited to, lysine (K), arginine (R) or histidine (H). The positively charged amino acids may also include positively charged unnatural amino acids such as ornithine; 2,4-diaminobutanoic acid 2,3-diaminopropanoic acid), 3-(aminoiminomethyl)amino-alanine, 2-amino-4-(aminoiminomethyl)aminobutanoic acid, N6-(aminoiminomethyly) lysine, 2-amino-7-(aminoiminomethyl)aminoheptanoic acid, 2,7-diaminoheptanoic acid, 2,8-diaminooxtanoic acid, 2,9-diaminononanoic acid, 2,10-diaminodecanoic acid, 4-(aminoiminomethyl)phenylalanine or 4-(aminoiminomethyl)aminophenylalanine.
Exemplary peptides represented by the consensus sequence Q1-L2-G3-X4-X5-X6-X7-X8-X9-R10-P11-S12-X13-X14-X15-R16-H17-W18 include, but are not limited to, the following sequences:
i)
(SEQ ID NO: 3)
QLGKKKHRRRPSKKKRHW;
ii)
(SEQ ID NO: 4)
QLGRRRHRRRPSRRRRHW;
iii)
(SEQ ID NO: 5)
QLGKKILAARPSKKKRHW;
iv)
(SEQ ID NO: 37)
HRRRPSKKKRHW;
and
v)
(SEQ ID NO: 38)
QLGAAAHRRRPSKKKRHW.
In one example, the isolated cytotoxic peptide as described herein excludes the sequence of QLGKKKHRRRPSKKKRHW (SEQ ID NO: 3).
In some examples, the isolated cytotoxic peptide comprising the amino acid sequence represented by the consensus sequence as described above can include one or more additional amino acids at either end of the amino acid sequence represented by the consensus sequence. For example, the isolated cytotoxic peptide can include at least 1, at least 3, at least 5, at least 7, at least 9, at least 11, at least 13, at least 15, at least 17, at least 19, or 2, 4, 6, 8, 10, 12, 14, 16, 18, 20 additional amino acid residues at the N-terminal and/or the C-terminal of the amino acid sequence represented by the consensus sequence.
In some examples, the functional part of the amino acid sequence represented by the consensus sequence comprises at least the following stretches of amino acid residues X8-X9-R10 and X13X14X15. Exemplary peptides comprising such functional part include, but are not limited to, the following sequences:
i)
(SEQ ID NO: 37)
HRRRPSKKKRHW;
and
ii)
(SEQ ID NO: 38)
QLGAAAHRRRPSKKKRHW.
In some other examples, the isolated cytotoxic peptide comprising the amino acid sequence represented by the consensus sequence as described above can include more than one amino acid sequence represented by the consensus sequence. In some specific examples, the isolated cytotoxic peptide as described above includes at least 2, at least 3 or at least 4 amino acid sequences represented by the consensus sequence. In some examples, at least 2 or at least 3 of the amino acid sequences represented by the consensus sequence are of the same sequence. In some other examples, all the amino acid sequences represented by the consensus sequence are different. In some examples, the more than one amino acid sequences represented by the consensus sequence are separated by 1 to 3, 4 to 6, 7 to 9, 10 to 12, 11 to 13, 14 to 16, 17 to 19, 20 to 22 or 23 to 25 other amino acid residues. In some other examples, at least 2 or at least 3 of the more than one amino acid sequences represented by the consensus sequence are not separated by any other amino acid residues.
In some examples, the isolated cytotoxic peptide as described herein can exert it cytotoxicity via the apoptosis-independent pathway, the p53-independent pathway or the p-TEFb-independent pathway. Thus, in some examples, the isolated cytotoxic peptide or conjugate thereof is effective against cancer resistant to p53-directed therapy, cancer resistant to the TEFb-directed pathway, or cancer resistant to therapies targeting other apoptosis-dependent pathways.
The term “resistant” or grammatical variants are used herein in the context of being drug-resistant, and refer to the reduction in effectiveness of a drug in curing a disease or condition. Drug resistance can result from, for example, defects in a pathway or defects in element(s) in a pathway targeted by the drug. In one specific example, a patient with a mutated or an otherwise defective p53 protein will show resistance to a drug targeting the p53-dependent pathway.
The isolated cytotoxic peptide as disclosed herein may contain peptide modifications. The term “modification” is used herein to refer to post-translational modification in general, which are modifications that occur on a peptide usually after its translation by ribosome is complete. Post-translational modification generally refers to the addition of a functional group covalently to a protein. These modifications include, but are not limited to, phosphorylation, glycosylation, sulfation, biotinylation, hydroxylation, acetylation, ubiquitination, nitrosylation, methylation, acetylation and lipidation.
In some examples, the isolated cytotoxic peptide as described herein contains at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, or 1 to 4, 5 to 8, 9 to 12, 13 to 16, 17 to 20, 21 to 24, 25 to 28, 29 to 32, 33 to 36, 37 to 40, 41 to 44, 45 to 48 or 49 to 52 modified amino acid residues. In some examples, all the modified amino acid residues contain the same type of modification. In some other examples, at least some of the modified amino acid residues contain different types of modifications. In some examples, wherein the isolated cytotoxic peptide comprises more than one amino acid sequence represented by the consensus sequence Q1-L2-G3-X4-X5-X6-X7-X8-X9-R10-P11-S12-X13-X14-X15-R16-H17-W18 (SEQ ID NO: 36), at least two of the amino acid sequences represented by the consensus sequence contain the same amino acid modifications. In some other examples, wherein the isolated cytotoxic peptide comprises more than one amino acid sequence represented by the consensus sequence, all the amino acid sequences represented by the consensus sequence are modified differently. In some examples, one amino acid residue may contain multiple modifications, such as one, two or three modifications. In some examples, all the modifications on the same amino acid residue are of the same type. In some other examples, at least some of the modifications on the same amino acid residue are of different types. Examples of such modification include, but are not limited to glycosylation, sulfation, phosphorylation, ubiquitination, methylation, lipidation, biotinylation, hydroxylation and acetylation.
The term “glycosylation” as used herein refers to the addition of a glycosyl group, usually to, but not limited to an arginine, an asparagine, a cysteine, a hydroxylysine, a serine, a threonine, a tyrosine, or a tryptophan residue, resulting in a glycoprotein. A glycosyl group refers to a substituent structure obtained by removing the hemiacetal hydroxyl group from the cyclic form of a monosaccharide and, by extension, of a lower oligosaccharide.
The term “sulfation” as used herein refers to the addition of a sulfo group usually to a tyrosine residue. A sulfo group refers to group SO3H, derived from sulfuric acid.
The term “phosphorylation” as used herein refers to the addition of a phosphate group. Protein phosphorylation commonly takes place at the serine, threonine, tyrosine, arginine, lysine, or cysteine residues. Phosphorylation may alter the structural conformation of a protein, causing it to become activated, deactivated, or modifying its function.
The term “ubiquitination” as used herein refers to the addition of a ubiquitin. Ubiquitin is a small protein that is found in almost all cellular tissues in humans and other eukaryotic organisms, which helps to regulate the processes of other proteins in the body. The genes in the human genome that produce ubiquitin include, but are not limited to: UBB, UBC, UBA52 and RPS27A. Ubiquitination may affect, for example, the activity and location of a protein, as well the interaction of the ubiquitinated protein with other proteins. In some examples, ubiquitination may lead to the degradation of the protein. In some examples, the protein is degraded via the proteasome.
The term “methylation” as used herein refers to the addition of a methyl group. A methyl group refers to an alkyl derived from methane, containing one carbon atom bonded to three hydrogen atoms, that is —CH3. Protein methylation can commonly take place at the arginine or lysine amino acid residues.
The term “lipidation” as used herein refers to the addition of a lipid group. Examples of lipidation include but are not limited to N-Myristoylation, Palmitoylation, Glycosylphosphatidylinositol (GPI)-anchor addition and Prenylation.
The term “biotinylation” as used herein refers to the addition of a biotin. A biotin refers to 5-[(3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl]pentanoic acid, with the following structural formula:
Figure US10500288-20191210-C00001
The term “hydroxylation” as used herein refers to the addition of a hydroxyl group (—OH).
The term “acetylation” as used herein refers to the addition of an acetyl group. An acetyl group contains a methyl group single-bonded to a carbonyl, represented by the following formula:
Figure US10500288-20191210-C00002
Amino acid changes in the polypeptide or fragment thereof may be effected by techniques well known to persons skilled in the relevant art. For example, amino acid changes may be effected by nucleotide replacement techniques which include the addition, deletion or substitution of nucleotides, under the proviso that the proper reading frame is maintained. Exemplary techniques include random mutagenesis, site-directed mutagenesis, oligonucleotide-mediated or polynucleotide-mediated mutagenesis, deletion of selected region(s) through the use of existing or engineered restriction enzyme sites, and the polymerase chain reaction.
The cell internalization of the cytotoxic peptide disclosed herein may require facilitators such as cell penetrating or cell targeting peptides or proteins. Thus, in one example, the isolated cytotoxic peptide as described herein is conjugated to at least one internalizing peptide or protein.
The term “conjugate” as used herein refers to the attachment of the isolated cytotoxic peptide to another object, such as other peptide(s) or polypeptide(s), protein(s), antibody(ies), or particle(s). The attachments can be resulted from covalent bonding or weak interactions. The term “conjugation” should be construed accordingly.
In some examples, the isolated cytotoxic peptide as described herein can be conjugated to an antibody.
The term “antibody” is used in the broadest sense to refer to an immunoglobulin molecule, a fragment of an immunoglobulin molecule, or a derivative of either thereof. The antibody can be a monoclonal antibody or a polyclonal antibody. In some examples, the antibody is an antibody for cancer therapy. Examples of an antibody for cancer therapy include, but are not limited to, Ado-trastuzumab emtansine (CAS No. 1018448-65-1), Alemtuzumab (CAS No. 216503-57-0), Bevacizumab (CAS No. 216974-75-3), Blinatumomab (CAS No. 853426-35-4), Brentuximab vedotin (CAS No. 914088-09-8), Cetuximab (CAS No. 205923-56-4), Daratumumab (CAS No. 945721-28-8), Denileukin diftitox (CAS No. 173146-27-5), Gemtuzumab (CAS No. 220578-59-6), Ibritumomab tiuxetan (CAS No. 174722-31-7), Ipilimumab (CAS No. 477202-00-9), Nivolumab (CAS No. 946414-94-4), Ofatumumab (CAS No. 679818-59-8), Panitumumab (CAS No. 339177-26-3), Rituximab (CAS No. 174722-31-7), Tositumomab (CAS No. 192391-48-3) and Trastuzumab (CAS No. 180288-69-1). Other antibodies for cancer therapy can also be used.
The isolated cytotoxic peptide as described herein can be conjugated to a protein or an antibody by, for example but not limited to, a cross-linker, a bifunctional linker or a trifunctional linker. Linker groups can be, for example but not limited to, small organic compounds or peptides substituted with chemical linkers. In some examples, the chemical linkers can be, but not limited to, any one of the following: succinimidyl 3-(2-pyridyldithio)propionate (SPDP), succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC), succinimidyl 4-(p-maleimidophenyl)butyrate (SMPB), 4-succinimidyloxycarbonyl-alpha-methyl-α(2-pyridyldithio)toluene (SMPT), EMCS (N-ε-malemidocaproyl-oxysuccinimide ester) and Sulfo-MBS (m-maleimidobenzoyl-N-hydroxysulfosuccinimide ester).
In some examples, a thiol group on the peptide is required for conjugating the isolated cytotoxic peptide as described herein to a linker. Thus, the isolated cytotoxic peptide will have at least one additional cysteine residue at either end of the peptide. Therefore, in some examples, the isolated cytotoxic peptide as described herein further comprises at least one cysteine residue at the N-terminal and/or the C-terminal end.
In general, the isolated cytotoxic peptide as described herein can exert its cytotoxic effect on any cell type, once the isolated cytotoxic peptide is internalized into the cell. Therefore, depending on the type of internalizing peptide or protein being conjugated to, the isolated cytotoxic peptide as described herein can exert its cytotoxic effect on a specific type of cell. In some examples, the internalizing peptide or protein is a cell penetrating peptide or protein. In some other examples, the internalizing peptide or protein is a cell targeting peptide or protein. In some examples, a cell penetrating peptide or protein can also function as a cell targeting peptide or protein. Examples of peptides that can function as both a cell penetrating peptide and a cell targeting peptide include but are not limited to, LTVSPWY peptide (LTV, SEQ ID NO: 8), HLYVSPW peptide (Pep2, SEQ ID NO: 795), CGFYWLRSC peptide (NRP, SEQ ID NO: 796) and CQDGRMGFC peptide (BLA, SEQ ID NO: 797). In some specific examples, the cell targeting peptide or protein is a cancer cell targeting peptide or protein. In some other specific examples, the cell targeting peptide or protein is a lipocyte targeting peptide or protein. In some further examples, the cell targeting peptide or protein is a stem cell targeting peptide or protein. In some examples, such conjugation can result in the internationalization of both the cytotoxic peptide and the internalizing peptide or protein. Examples of a cell-penetrating peptide include, but are not limited to, transactivator of transcription (TAT), penetratin, R6-Pen, transportan, MPG peptide, sweet arrow peptide (SAP), peptide from vascular endothelial-cadherin protein (pVEC), Pep-1 (KETWWETWWTEWSQPKKKRKV)(SEQ ID NO: 6), polylysines, polyarginines, model amphipathic peptide (MAP), FGF (AAVALLPAVLLALLAP) (SEQ ID NO: 798) and R6W3 (RRWWRRWRR)(SEQ ID NO: 7). Examples of a cancer cell-targeting peptide include but are not limited toarginine-glycine-aspartic acid (RGD), asparagine-glycine-arginine (NGR), TCP-1 phage peptide (TCP-1), and the peptides listed in Table 1. Examples of a lipocyte-targeting peptide include but are not limited to, CKGGRAKDC peptide (ATS, SEQ ID NO: 39). Examples of stem cell targeting peptide are listed in Table 2. Examples of conjugated peptides and their effects are discussed in details in Examples 6 to 8.
TABLE 1
Sequences of cancer cell targeting peptides
SEQ ID
NO: Cancer cell targeting peptide
  8 LTVSPWY
  9 IFLLWQR
 10 CGNSNPKSC
 11 SVSVGMKPSPRP
 64 CSNIDARAC
 65 CDPSRGKNC
 66 CPSDLKDAC
 67 CRTTRGTKC
 68 CRMTRNKPC
 69 CRVSRQNKC
 70 CAKIDPELC
 71 CGGERGKSC
 72 YSINDWH
 73 YSFNSWM
 74 PNPNNST
 75 YPTPYDI
 76 LPAMPNS
 77 CNRRTKAGC
 78 SRHDLNS
 79 STVATSQ
 80 QRLGNQWAVGHLM
 81 RGDF
 82 KGVSLSYR
 83 QFPPKLTNNSML
 84 SYDILKPNPQRL
 85 SHGKPPSFSPYT
 86 LLADTTHHRPWT
 87 CTPSPPFSHC
 88 CPNGRC
 89 CLSYYPSYC
 90 RTRYED
 91 GMMYRS
 92 RWRTNF
 93 RLQLKL
 94 RIPLEM
 95 QFDEPR
 96 TSAVRT
 97 GLWQGP
 98 QCTGRF
 99 LPGMMG
100 DVGTTE
101 TDLGAM
102 DSNAES
103 ITDMAA
104 WRPCES
105 WRNTIA
106 IDKQLE
107 FMEIET
108 HEVVAG
109 GGHTRQ
110 INGKVT
111 VPWXEPAYQRFL
112 GRDS
113 RGEPAYQRFL
114 RGDPAYQRFL
115 WXEPAYQGRFL
116 WXEPAYNGRFL
117 RGEPAYQGRFL
118 RGDPAYQGRFL
119 RGEPAYNGRFL
120 RGDPAYNGRFL
121 WXEPAYQRFL
122 AXEPAYQRFL
123 WAEPAYQRFL
124 WXAPAYQRFL
125 WXEAAYQRFL
126 WXEPAAQRFL
127 WXEPAYARFL
128 WXEPAYQAFL
129 WXEPAYQAAL
130 WXEPAYQAFA
131 EXEPAYQRFL
132 LXEPAYQRFL
133 KXEPAYQRFL
134 QXEPAYQRFL
135 YXEPAYQRFL
136 FXEPAYQRFL
137 F*XEPAYQRFL
138 WXEPAYQREL
139 WXEPAYQRLL
140 WXEPAYQRKL
141 WXEPAYQRQ
142 WXEPAYQRYL
143 WXEPAYQRF*L
144 WXEPAYQRFL
145 WXEPAYQRRE
146 WXEPAYQRFK
147 WXEPAYQRFQ
148 WXEPAYQRFT
149 WXEPAYQRFF
150 XEPAYQRFL
151 XEPAYQREL
152 XEPAYQRLL
153 XEPAYQRKL
154 XEPAYQRQL
155 XEPAYQRYL
156 XEPAYQRF*L
157 XEPAYQRFL
158 XEPAYQRFE
159 XEPAYQRFK
160 XEPAYQRFQ
161 XEPAYQRFT
162 XEPAYQRFF
163 KSLSRHDHIHHH
164 GGCLQILPTLSECFGR
165 GLKVCGRYPGICDGIR
166 GKYTWYGYSLRANWMR
167 VPCQKRPGWVCLW
168 KWCVIWSKEGCLF
169 SSWCMRGQYNKICMW
170 VECYLIRDNLCIY
171 WWCLGERVVRCAH
172 FYCVIERLGVCLY
173 RVCFLWQDGRCVF
174 NRLKCRAQATHSAAPCIRGY
175 RQNSCTYSDARRWALCWSGE
176 QLNSCIFISGDRAIRGCMDWV
177 KYGLCRDETVFPSHSCTFTG
178 GSPQCPGGFNCPRCDCGAGY
179 GTGSCGYGKLHTGYWCSYFP
180 NSSSCDTSVVRSTWACILQP
181 VRAVCTTLKSRGHEECWSLQ
182 VYAQCGVNVRTGRGGCSRLM
183 VHMNCSWMRVSEGHPCESAD
184 GRQGCYEHLWRLIAWCAIFL
185 LRMTCAFGVAQRSADCALSS
186 SIVNCSAALTDLPTRCGGNI
187 CGTRCVRCQNGPEASCEQPL
188 TPLFCGNHGRQPSPLCMKWD
189 FTTVCRQPRGHEAIVCGSGK
190 APSFCGTAMLGASRYCYSGP
191 GARECESGGPGMRKLCTQIN
192 NNRACFRTSKGNPAECPYLG
193 GSLACQNIVVCVKKQCNALC
194 KRASCQNPLFSNFFVCGLSE
195 LPNFCMDTSGRAGPLCMGSE
196 RHTVCRVSLSSVQGSCSHEY
197 CGLIIQKNEC
198 CNAGESSKNC
199 GRRTRSRRLRRS
200 SMSIASPQIPWS
201 TPRNLRTSNTHR
202 GRRIAGPYIALE
203 SMPINSPYIPWS
204 GRRPMKLNKTP
205 GRRINTRLILPRN
206 GRRTRSSRLRNS
207 CLSDGKRKC
208 CLDGGRPKC
209 CREAGRKAC
210 CAGRRSAYC
211 CNRRTKAGC
212 CPIEDRPMC
213 CGRRAGGSC
214 CGNSNPKSC
215 CPHNLTKLC
216 GPLPLR
217 CDCRGDCFC
218 GHGKHKNK
219 HKHGHGHGKHKNKGK
220 KHGHGHGK
221 KGHHGKHG
222 HKNKGKKN
223 CRGRRST
224 CRSRKG
225 CKAAKNK
226 CKGAKAR
227 FRVGVADV
228 CEYQLDVE
229 CSRPRRSEC
230 CGKRK
231 CDTRL
232 CGTKRKC
233 CDTAVVEGL
234 CRSRKG
235 CEYQLDVE
236 CPIEDRPMC
237 PIEDRPM
238 PIDERPM
239 ALRDRPM
240 PMMRQRPM
241 PLASRPM
242 PEKFRPM
243 VPEQRPM
244 DLPMHPM
245 QFQSQPM
246 QPPMEYS
247 NGRSL
248 MTQMIS
249 TALSPQ
250 WNLPWYYSVSPT
251 LTVLPW
252 LTVEPWL
253 LTVSPLWD
254 LTVTPWL
255 LTVQPWP
256 LTVSPWT
257 VLTVQPW
258 LTVSLWT
259 PGVIPWN
260 LTYQTWP
261 ELYVSRL
262 NLYYASW
263 TLTVLPW
264 NLYVASW
265 SMSIARL
266 VSFLEYR
267 CPGPEGAGC
268 RGDfK
269 PRPGAPLAGSWPGTS
270 ADGAPRPGAPLA
271 DRWRPALPVVLFPLH
272 ASSSYPLIHWRPWAR
273 DRWRPALP
274 IHWRPWAR
275 AAEWLDAFFVRHVDR
276 GDVWLFLTSTSHFAR
277 GCSVSSVGALCTHV
278 APCCSHLDASPFQRP
279 AQSNFVTWGYNVAV
280 RASDVGSDVVPRYPF
281 MARSGL
282 MARAKE
283 MSRTMS
284 MTKSAG
285 MTKCRG
286 MTRNLQ
287 MTRQIG
288 MSRPHK
289 MAKHAM
290 CWWRLEGC
291 CLQLFSTC
292 CAKGYRSC
293 CTGSWLGC
294 AEGEFMYWGDSHWLQYWYEGDPAK
295 AEGEFWGDSHWLQYWYEGDPAK
296 AEGEFIHNRYNRFFYWYGDPAK
297 AEGEFPRWGDSHWLQYWYEGDPAK
298 AEGEFLMWGGSHWLEYWYEGDPAK
299 AEGEFGHWCDQHWLQYWYEGDPAK
300 AEGEFGWWGDSHWLQYEGDPAK
301 CRGDCF
302 CDCRGDCFC
303 CNGRCVSGCAGRC
304 CGSLVRC
305 CRGDCGGKWCFRVCYRGICYRRCR
306 MCPKHPLGC
307 LCPKHPLGC
308 HLQIQPWYPQIS
309 VPWMEPAYQRFL
310 LSSVNSFPVVTP
311 QPWLEQAYYSTF
312 SALLPWPVLVNY
313 ITTPWDEMRSFL
314 HSFLHPWDLFDY
315 VPWMEPAYQRFL
316 MLPKPSSFPVPG
317 HSFLHPWDLFDY
318 CNGRCVSGCAGRC
319 CVLNGRMEC
320 FDDARL
321 FSDARL
322 FSDMRL
323 FVDVRL
324 FTDIRL
325 FNDYRL
326 FSDTRL
327 DPAFIFYHSTLFFNS
328 GGHDGDPVLTGTLFY
329 AVDPRMFYLLLRGGA
330 PIHYIF
331 YIHYIF
332 RIHYIF
333 WREWFL
334 WWAMKP
335 LILSSGELLRHPRG
336 TAASGVRSMH
337 LTLRWVGLMS
338 GGGTRAGMKY
339 WGKIEDPLRA
340 AGQTLTASGD
341 DLLAVSWLRA
342 SAERGVVAMS
343 AIHSELMWVS
344 FWTERAGWAY
345 MVWSKGPLFL
346 AGTRMSWEVL
347 VSRSSRWGSI
348 DAHVLVPRTP
349 AQGIVLQLAL
350 LSPLLSPATA
351 CDCRGDCFC
352 CNGRCVSGCAGRC
353 NGRAHA
354 CVLNGRMEC
355 HGRFILPWWYAFSPS
356 RFRGLISLSQVYLSP
357 ARVSFWRYSSFAPTY
358 GSWYAWSPLVPSAQI
359 KKEKDIMKKTI
360 GRGDSPK
361 SNPFSKPYGLTV
362 YPHYSLPGSSTL
363 KDEPQRRSARLSAKPAPPKPEPKPKKAPA
KK
364 TLTYTWS
365 CREKA
366 CGQKRTRGC
367 RPARPAR
368 CRGDGWC
369 RGDGWK
370 TSPLNIHNGQKL
371 CRGDKGPDC
372 CRGDRGPDC
373 CRGDKTTNC
374 CRGDHAGDC
375 CRGDHGVEC
376 CGRGDNLPC
377 CGRGDNLAC
378 CEKRGDNLC
379 CEKRGDSVC
380 CSGRGDSLC
381 CGKRGDSIC
382 CTGRGDALC
383 CRGDSAC
384 CRGDKGENC
385 CGRGDSPDC
386 CRGDKGPEC
387 CRGDKHADC
388 CRGDHAANC
389 CRGDAGINC
390 CGRGDMPSC
391 CEKRGDSLC
392 KDEPQRRSARLSAKPAPPKPEPKPKKAPA
KK
393 CREKA
394 TDSILRSYDWTY
395 DMPKQLLAPWYY
396 DMPKQLLAPWYY
397 SYPLSFLGPLIS
398 TQQPLEGHQLPY
399 TGVSWSVAQPSF
400 SVSVGMKPSPRP
401 SQWNSPPSSAAF
402 CGNSNPKSC
403 SFSIIHTPILPL
404 GNGRAHA
405 AHLPIVRASLPS
406 TPMNHHSQHAER
407 GNIIPDRPMHPT
408 FPSSLIIPPLPN
409 EDIKPKTSLAFR
410 YEDIKPKTSLAFR
411 TQPADLQTHNHN
412 FDHSSKWTRTSP
413 YSHNTITNLYFS
414 WPRYAESTLQLR
415 KGVSLSYRKKGVSLSYR
416 SVSVGMKPSPRP
417 WPLHTSVYPPSP
418 NTLPPFSPPSPP
419 SFPDSNIAPSSP
420 QHAPSNSKSVLT
421 WPTYLNPSSLKA
422 GPSGNLHIRPAS
423 SPLLSTRAVQLS
424 SPMFTMIQGDAQ
425 VNSHQALWSPAQ
426 STLPPPLRFANV
427 SFNQPYLYKTAF
428 YHTRIALPDNLP
429 AQSTAFQKPLLM
430 KCCYSL
431 RLLDTNRPLLPY
432 CSDSWHYWC
433 FQHPSFI
434 SMSIASPYIALE
435 SMSIASPY1PWS
436 SPGPMKLLKTPL
437 TLNINRLILPRT
438 SMSIGSPYITFG
439 VPNTNSLPAAVN
440 LIAKTALPQTNK
441 LIAKTALPQTN
442 CPHSKPCLC
443 GGCRGDMFGC
444 FRPNRAQDYNTN
445 QEFSPYMGLEFKKH
446 QEFSPNLWGLEFQKN
447 QEYSPNLWGHEFRSH
448 HTFEPGV
449 PSTNHAL
450 PSTLTSS
451 APSQTYH
452 KAMSWYA
453 SRESPHP
454 QSRLSLG
455 LDHFAPM
456 LDKKTTS
457 NMSPQLD
458 SQRQTLD
459 STKLLHE
460 TSPTNRS
461 PHSPTSL
462 HGKYFVS
463 PQRHVNY
464 MMSQLAH
465 PMAHLEF
466 ELIKESR
467 QPENLPT
468 NTHMTAF
469 PFKLSKH
470 ASSLHTI
471 HPLRLPA
472 HQSVNKE
473 LQNPTPE
474 PTEAQLQ
475 LFAQLGP
476 NQPTRAL
477 TPRTQKA
478 IHFPSAS
479 PLRIAQH
480 CRLTGGKGVGC
481 CRRTNWQGAGC
482 CQLTGTHGAGC
483 CADPNSVRAMC
484 CADPNSVRAHC
485 CAAHYRVGPWC
486 PQNSKIPGPTFLDPH
487 SMEPALPDWWWKMFK
488 DKPTAFVSVYLKTAL
489 APRPGPWLWSNADSV
490 GVTDSSTSNLDMPHW
491 PKMTLQRSNIRPSMP
492 LYPLHTYTPLSLPLF
493 LTGTCLQYQSRCGNTR
494 AYTKCSRQWRTCMTTH
495 ANTPCGPYTHDCPVKR
496 NISRCTHPFMACGKQS
497 PRNICSRRDPTCWTTY
498 GCNGRC
499 QHWSYKCIRP
500 CVSNPRWKC
501 CHVLWSTRC
502 SWLAYPGAVSYR
503 YSAYPDSVPMMS
504 SRESPHP
505 VPWMEPAYQRFL
506 CDSDSDITWDQLWDLMK
507 ATLDGVS
508 RRHSVSG
509 SGWFAGS
510 GSVSHRR
511 GSVLPVL
512 RSGRVSN
513 NSVRGSR
514 NVVRQ
515 CDCRGDCFC
516 CSDSWHYWC
517 CSDWQHPWC
518 CSDYNHHWC
519 CSDGQHYWC
520 CYDSWHYWC
521 CFDGNHIWC
522 CTDFPRSFC
523 CTQDRQHPC
524 CLSRYLDQC
525 CPRECESIC
526 CTTHWGFTLC
527 CRRHWGFEFC
528 CVPELGHEC
529 HTMYYHHYQHHL
530 CGNKRTRGC
531 CGRECPRLCQSSC
532 CGEACGGQCALPC
533 IWSGYGVYW
534 PSCAYMCIT
535 WESLYFPRE
536 SKVLYYNWE
537 CGLMCQGACFDVC
538 CERACRNLCREGC
539 CPRGCLAVCVSQC
540 CKVCNGRCCG
541 CEMCNGRCMG
542 CPLCNGRCAL
543 CPTCNGRCVR
544 CGVCNGRCGL
545 CEQCNGRCGQ
546 CRNCNGRCEG
547 CVLCNGRCWS
548 CVTCNGRCRV
549 CTECNGRCQ
550 CRTCNGRCLE
551 CETCNGRCVG
552 CAVCNGRCGF
553 CSCCNGRCGD
554 CWGCNGRCRM
555 CPLCNGRCAR
556 CKSCNGRCLA
557 CVPCNGRCHE
558 CQSCNGRCVR
559 CRTCNGRCQV
560 CVQCNGRCAL
561 CRCCNGRCSP
562 CASNNGRVVL
563 CGRCNGRCLL
564 CWLCNGRCGR
565 CSKCNGRCGH
566 CVWCNGRCGL
567 CIRCNGRCSV
568 CGECNGRCVE
569 CEGVNGRRLR
570 CLSCNGRCPS
571 CEVCNGRCAL
572 GRSQMQI
573 HHTRFVS
574 SKGLRHR
575 VASVSVA
576 WRVLAAF
577 KMGPKVW
578 IFSGSRE
579 SPGSWTW
580 NPRWFWD
581 GRWYKWA
582 IKARASP
583 SGWCYRC
584 ALVGLMR
585 LWAEMTG
586 CWSGVDC
587 DTLRLRI
588 SKSSGVS
589 IVADYQR
590 VWRTGHL
591 VVDRFPD
592 LSMFTRP
593 GLPVKWS
594 IMYPGWL
595 CVMVRDGDC
596 CVR1RPC
597 CQLAAVC
598 CGVGSSC
599 CVSGPRC
600 CGLSDSC
601 CGEGHPC
602 CYTADPC
603 CELSLISKC
604 CPEHRSLVC
605 CLVVHEAAC
606 CYVELHC
607 CWRKFYC
608 CFWPNRC
609 CYSYFLAC
610 CPRGSRC
611 CRLGIAC
612 CDDSWKC
613 CAQLLQVSC
614 CYPADPC
615 CKALSQAC
616 CTDYVRC
617 CGETMRC
618 CLSGSLSC
619 WGTGLC
620 GICKDDWCQ
621 TSCDPSLCE
622 KGCGTRQCW
623 YRCREVLCQ
624 CWGTGLC
625 WSCADRTCM
626 AGCRLKSCA
627 SRCKTGLCQ
628 PICEVSRCW
629 WTCRASWCS
630 GRCLLMQCR
631 TECDMSRCM
632 ARCRVDPCV
633 CIEGVLGGC
634 CSVANSC
635 CSSTMRC
636 SIDSTTF
637 GPSRVGG
638 WWSGLEA
639 LGTDVRQ
640 LVGVRLL
641 GRPGDJW
642 TVWNPVG
643 GLLLVVP
644 FAATSAE
645 WCCRQFN
646 VGFGKAL
647 DSSLRLP
648 KLWCAMS
649 SLVSFLG
650 GSFAFLV
651 IASVRWA
652 TWGHLRA
653 QYREGLV
654 QSADRSV
655 YMFWTSR
656 LVRRWYL
657 TARGSSR
658 TTREKNL
659 PKWLLFS
660 LRTNVVH
661 AVMGLAA
662 VRNSLRN
663 TDCTPSRCT
664 SWCQFEKCL
665 VPCRFKQCW
666 CTAMRNTDC
667 CRESLKNC
668 CMEMGVKC
669 VTCRSLMCQ
670 CNNVGSYC
671 CGTRVDHC
672 CISLDRSC
673 CAMVSMED
674 CYLGVSNC
675 CYLVNVDC
676 CIRSAVSC
677 LVCLPPSCE
678 RHCFSQWCS
679 FYCPGVGCR
680 ISCAVDAC
681 EACEMAGCL
682 PRCESQLCP
683 RSCIKHQCP
684 QWCSRRWCT
685 MFCRMRSCD
686 GICKDLWCQ
687 NACESAICG
688 APCGLLACI
689 NRCRGVSCT
690 FPCEGKKCL
691 ADCRQKPCL
692 FGCVMASCR
693 AGCINGLCG
694 RSCAEPWCY
695 DTCRALRCN
696 KGCGTRQCW
697 GRCVDGGCT
698 YRCIARECE
699 KRCSSSLCA
700 ICLLAHCA
701 QACPMLLCM
702 LDCLSELCS
703 AGCRVESC
704 HTCLVALCA
705 IYCPGQECE
706 RLCSLYGCV
707 RKCEVPGCQ
708 EDCTSRFCS
709 LECVVDSCR
710 EICVDGLCV
711 RWCREKSCW
712 FRCLERVCT
713 RPCGDQACE
714 CNKTDGDEGVTC
715 PQRRSARLSA
716 IELLQAR
717 ISLLQAR
718 IDLMQAR
719 IILLQGR
720 ISLLGAR
721 FSLLDAR
722 CTPSPFSHC
723 AGTRMSWEV
724 CSNRDARRC
725 WQPDTAHHWATL
726 HNAYWHWPPSMT
727 GHLIPLRQPSH
728 YTSPHHSTTGHL
729 WTHHHSYPRPL
730 NSFPLMLMHHHP
731 KHMHWHPPALN
732 SLDSMSPQWHAD
733 SEFIHHWTPPPS
734 NGFSHHAPLMRY
735 HHEWTHHWPPP
736 AWPENPSRRPF
737 AGFQHHPSFYRF
738 QRSPMMSRIRLP
739 YRHWPIDYPPP
740 MIHTNHWWAQD
741 CALIIQKNEC
742 CGLILQKNEC
743 CGLIIQRNEC
744 CGLIINKNEC
745 CNAAESSKNC
746 CNAGESSRNC
747 CNAGESTKNC
748 CNAGDSSKNC
749 CLSDGK
750 CLSDGKPVS
751 CSMSAKKKC
752 CKTRVSCGV
753 CASLSCR
754 CSGGKVLDC
755 CASLSCR
756 CSGGKVLDC
757 CSMSAKKKC
758 CKTRVSCGV
759 CASLSCR
760 WIFPWIQL
761 WDLAWMFRLPVG
762 CRGSGAGRC
763 CKGGRAKDC
764 CGSPGWVRC
765 VGVGEWSV
766 SRPRR
767 CXSRPRRZC
768 CSRPRRSVC
769 CSRPRRSWC
770 CGLSGLGVA
771 CPIRPMEDC
772 CPIDERPMC
773 CALRDRPMC
774 CPEKFRPMC
775 CSPQSQPMC
776 CGLIIQKNEC
777 CRGDK
778 KDEPQRRSARLSAKPAPPKPEPKPKKAPA
KK
779 CGQKRTRGC
780 NVVRQ
781 PHSCNK
782 HPLSKHPYWSQP
783 IFLLWQR
784 APRPG
785 CRGDRCPDC
786 CVNHPAFAC
787 KDEPQRRSARLSAKPAPPKPEPKPKKAPA
KK
788 CGKRK
789 KDEPQRRSARLSAKPAPPKPEPKPKKAPA
KKC
TABLE 2
Sequences of stein cell targeting peptides
SEQ ID NO: Stem cell targeting peptide sequence
41 AVGPYSGLKVFR
42 SPLSKGSTHLST
43 TNKPVQPRQTLP
44 AMHPSPASAKMN
45 AGAPYRNTNAGT
46 TPTERFNTHHVE
47 QAGDEKEWLGPK
48 GSNQSVRYLQQT
49 NVDYAFGKREQS
50 DPLLHSQADVQS
51 LPHSSWNPKLAL
52 HQVHAKPLDLMP
53 QYTFSVNPLMRA
54 VPHFSTPTSVFQ
55 NYAIAVVNVLSH
56 NTKVPDPTARLL
57 HGAAWGTRTGHV
58 VPATETAQAGHA
59 VEYHFNHTMTAY
60 VGGEAWSSPTDL
61 HSDQVALKMTRS
62 QPHSVSVSDTWH
63 SNNDNLAHRVRL
The isolated cytotoxic peptide or conjugate thereof as described herein can exert its cytotoxicity selectively. Thus, in some examples, the isolated cytotoxic peptide or conjugate thereof has low toxicity towards cells and tissue that are not the target of the conjugated cell penetrating peptide or cell targeting peptide.
In some examples, the isolated cytotoxic peptide or conjugate thereof exerts its cytotoxic effects only upon internalization into a target cell.
The stability of the isolated cytotoxic peptide as disclosed herein can be enhanced by conjugating to a particle, in particular a small particle. Examples of a particle include but are not limited to, nanoparticle or microparticle. In some examples, the nanoparticle or microparticle is made from materials including but not limited to, metal, silica, carbon, polymeric materials, and mixtures thereof. Examples of metal include, but are not limited to, gold and silver. Examples of polymeric materials include, but are not limited to, bio-degradable polymers such as a poly(lactide-co-glycolide), poly(lactic acid), poly(alkylene glycol), polybutylcyanoacrylate, poly(methylmethacrylate-co-methacrylic acid), poly-allylamine, polyanhydride and polyhydroxybutyric acid.
In the present disclosure, there is provided an isolated nucleic acid molecule comprising the nucleotide sequence encoding a peptide as described herein.
Since the degeneracy of the genetic code permits substitutions of certain codons by other codons which specify the same amino acid and hence give rise to the same protein, the disclosure is not limited to a specific nucleic acid molecule but includes all nucleic acid molecules comprising a nucleotide sequence coding for the peptide as described herein.
A nucleic acid molecule, such as DNA (including cDNA and genomic DNA), RNA (such as mRNA), is regarded to be ‘capable of expressing a nucleic acid molecule or a coding nucleotide sequence’ or capable ‘to allow expression of a nucleotide sequence’ if it contains regulatory nucleotide sequences which contain transcriptional and translational information and such sequences are “operably linked” to nucleotide sequences which encode the peptide. An operable linkage is a linkage in which the regulatory DNA sequences and the DNA sequences sought to be expressed are connected in such a way as to permit gene sequence expression. The precise nature of the regulatory regions needed for gene sequence expression may vary from organism to organism, but shall, in general include a promoter region which, in prokaryotes, contains only the promoter or both the promoter which directs the initiation of RNA transcription as well as the DNA sequences which, when transcribed into RNA will signal the initiation of synthesis. Such regions will normally include non-coding regions which are located 5′ and 3′ to the nucleotide sequence to be expressed and which are involved with initiation of transcription and translation such as the TATA box, capping sequence and CAAT sequences. These regions can for example, also contain enhancer sequences or translated signal and leader sequences for targeting the produced peptide to a specific compartment of a host cell, which is used for producing a peptide described above. The isolated nucleic acid molecule as described herein can be obtained by cloning or produced synthetically. Examples of the isolated nucleic acid molecules are listed in Table 3.
TABLE 3
Examples of isolated nucleic acid molecules
encoding the polypeptides as described herein
SEQ
ID
NO: Sequences (5′ to 3′)
790 cagctgggcaaaaaaaaacatcgccgccgcccgagcaaaaaaaaa
cgccattgg
791 cagctgggcgcggcggcgcatcgccgccgcccgagcaaaaaaaaa
cgccattgg
792 cagctgggccgccgccgccatcgccgccgcccgagccgccgccgc
cgccattgg
793 cagctgggcaaaaaaattctggcggcgcgcccgagcaaaaaaaaa
cgccattgg
794 catcgccgccgcccgagcaaaaaaaaacgccattgg
The isolated nucleic acid molecule comprising the nucleotide sequence encoding the peptide as disclosed herein can be comprised in a vector, for example an expression vector. Thus, in the present disclosure, there is provided a vector comprising an isolated nucleic acid molecule as described herein. Representative vectors include plasmids, cosmids, and viral vectors. Vectors can also comprise nucleic acids including expression control elements, such as transcription/translation control signals, origins of replication, polyadenylation signals, internal ribosome entry sites, promoters and enhancers, wherein the control elements are operatively associated with a nucleic acid encoding a gene product. Selection of these and other common vector elements are conventional and many such sequences can be derived from commercially available vectors. A vector can be introduced into targeting cells using any suitable method known in the art for introducing DNA into cells, including but not limited to microinjection, electroporation, calcium phosphate precipitation, liposome-mediated delivery, viral infection, protoplast fusion, and particle-mediated uptake.
The vector comprising the isolated nucleic acid molecule can be transformed into host cells capable of expressing the genes. The transformation can be carried out in accordance with standard techniques. Thus, the disclosure is also directed to a (recombinant) host cell containing a vector as described herein. In this context, the transformed host cells can be cultured under conditions suitable for expression of the nucleotide sequence encoding the peptide as described above. Host cells can be established, adapted and completely cultivated under serum free conditions, and optionally in media which are free of any protein/peptide of animal origin. Commercially available media such as RPMI-1640 (Sigma), Dulbecco's Modified Eagle's Medium (DMEM; Sigma), Minimal Essential Medium (MEM; Sigma), CHO-S-SFMII (Invitrogen), serum free-CHO Medium (Sigma), and protein-free CHO Medium (Sigma) are exemplary appropriate nutrient solutions. Any of the media may be supplemented as necessary with a variety of compounds, examples of which are hormones and/or other growth factors (such as insulin, transferrin, epidermal growth factor, insulin like growth factor), salts (such as sodium chloride, calcium, magnesium, phosphate), buffers (such as HEPES), nucleosides (such as adenosine, thymidine), glutamine, glucose or other equivalent energy sources, antibiotics, trace elements. Any other necessary supplements may also be included at appropriate concentrations that are known to those skilled in the art.
The isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule or the vector as described herein can be formulated into compositions, for example pharmaceutical compositions, suitable for administration. Where applicable, the peptide or conjugate thereof, the nucleic acid molecule or the vector can be administered with a pharmaceutically acceptable carrier. A “carrier” can include any pharmaceutically acceptable carrier as long as the carrier is compatible with other ingredients of the formulation and not injurious to the patient. Accordingly, pharmaceutical compositions for use may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Thus, in one example, the present disclosure provides a pharmaceutical composition comprising, but not limited to, an isolated cytotoxic peptide or conjugate thereof, an isolated nucleic acid molecule or a vector as described herein. In another example, the pharmaceutical composition further comprises one or more pharmaceutically acceptable excipients, vehicles or carriers.
Examples of pharmaceutically acceptable excipients, carriers or diluents are demineralised or distilled water; saline solution; vegetable based oils such as peanut oil, safflower oil, olive oil, cottonseed oil, maize oil, sesame oils such as peanut oil, safflower oil, olive oil, cottonseed oil, maize oil, sesame oil, arachis oil or coconut oil; silicone oils, including polysiloxanes, such as methyl polysiloxane, phenyl polysiloxane and methylphenyl polysolpoxane; volatile silicones; mineral oils such as liquid paraffin, soft paraffin or squalane; cellulose derivatives such as methyl cellulose, ethyl cellulose, carboxymethylcellulose, sodium carboxymethylcellulose or hydroxypropylmethylcellulose; lower alkanols, for example ethanol or iso-propanol; lower aralkanols; lower polyalkylene glycols or lower alkylene glycols, for example polyethylene glycol, polypropylene glycol, ethylene glycol, propylene glycol, 1,3-butylene glycol or glycerin; fatty acid esters such as isopropyl palmitate, isopropyl myristate or ethyl oleate; polyvinylpyrolidone; agar; gum tragacanth or gum acacia, and petroleum jelly. Typically, the carrier or carriers will form from 10% to 99.9% by weight of the compositions.
The isolated cytotoxic peptide or conjugate thereof as described herein may be present in the compositions in any of a wide variety of forms. For example, two, three, four or more peptides or conjugates thereof may be merely mixed together or may be more closely associated through complexation, crystallization, or ionic or covalent bonding. The peptides or conjugates thereof can also be formulated as prodrugs.
The term “prodrug” as used herein refers to compounds that rapidly convert in vivo into pharmacologically active compounds. Suitable prodrugs can be made, for instance, by conjugating the isolated cytotoxic peptide as described herein to one or more protective peptides. Various linkers known in the art can be used in such conjugations, for example but not limited to valine-citrulline dipeptide linker. Such linkers can be cleavable, resulting in the release of the pharmacologically active compounds. For example, valine-citrulline dipeptide linker is cleavable by lysosomal cathepsin B, resulting in the release of the cytotoxic peptide.
The isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule and the vector as described herein can also be used in combination with one or more other therapeutic agents to achieve better results of treatment and/or to reduce potential side effects. Thus, in one example, the pharmaceutical composition further comprises one or more therapeutic agent. The isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule and/or the vector as described herein may be administered simultaneously, sequentially or separately from the one or more further therapeutic agent. By simultaneous is meant that the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule or the vector as described herein may be contained in the same pharmaceutical composition as the one or more further therapeutic agent, or they are contained in different pharmaceutical compositions but taken at the same time. By sequential is meant that the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule or the vector as described herein and the one or more further therapeutic agent are contained in different pharmaceutical compositions, and are administered one immediately after another. By separately is meant the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule or the vector as described herein and the one or more further therapeutic agent are contained in different pharmaceutical compositions, and are administered with a period of time apart. For example, they can be administered separately with about 12 hours, 10 hours, 8 hours, 6 hours, 4 hours, 2 hours, 1 hour, 30 minutes, or 15 minutes apart. In some examples, when the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule or the vector as described herein and the one or more further therapeutic agent are contained in different pharmaceutical compositions, they can be administered using different routes.
The one or more therapeutic agent can be therapeutic agent against cancer, therapeutic agent for weight loss, or therapeutic agent with other effects, including but not limited to, reducing inflammation, reducing infection and relieving pain. In one example, the one or more therapeutic agent is a therapeutic agent against cancer, including but not limited to, general chemotherapeutic agents such as alkylating agents, antimetabolites, anti-tumor antibiotics, topoisomerase inhibitors, mitotic inhibitors and corticosteroids, inhibitors of translation and transcription; immunotherapeutic agents such as antibodies, in particular monoclonal antibodies, cytokines and vaccines; and targeted therapeutic agents such as small molecule drugs. In another example, the one or more therapeutic agent is a therapeutic agent for weight loss, including but not limited to, benzphetamine, bupropion hydrochloride, diethylpropion, lorcaserin, methamphetamine, naltrexone hydrochloride, orlistat, phendimetrazine, phentermine and sibutramine.
The isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, the vector or the pharmaceutical composition as described herein can be administered in a number of ways depending upon whether local or systemic administration is desired and upon the area to be treated. For example, administration may be oral, intraadiposal, intraarterial, intraarticular, intracranial, intradermal, intralesional, intramuscular, intranasal, intraocularal, intrapericardial, intraperitoneal, intrapleural, intraprostatical, intrarectal, intrathecal, intratracheal, intratumoral, intraumbilical, intravenous, intravesicularl, intravitreal, liposomal, local, mucosal, enteral, parenteral, rectal, subconjunctival, subcutaneous, sublingual, topical, transbuccal, transdermal, vaginal, in crèmes, in lipid compositions, via a catheter, via a lavage, via continuous infusion, via infusion, via inhalation, via injection, via local delivery, via localized perfusion or any combination thereof. One skilled in the art will be able to identify the method to adapt the pharmaceutically active component to the characteristics suitable for the selected route of administration. Such characteristics include, but are not limited to, solubility, stability, and deliverability.
Compositions and formulations for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets or tablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders may be desirable.
Compositions and formulations for parenteral, intrathecal or intraventricular administration may include sterile aqueous solutions that may also contain buffers, diluents and other suitable additives such as, but not limited to, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients.
Compositions as described herein include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions may be generated from a variety of components that include, but are not limited to, preformed liquids, self-emulsifying solids and self-emulsifying semisolids.
The formulations as described herein, which may conveniently be presented in unit dosage form, may be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredients with the pharmaceutical carrier(s) or excipient(s). In general the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
The compositions as described herein may be formulated into any of many possible dosage forms including, but not limited to tablets, capsules, liquid syrups and soft gels. The compositions as described herein may also be formulated as suspensions in aqueous, non-aqueous or mixed media. Aqueous suspensions may further contain substances that increase the viscosity of the suspension including, for example, sodium carboxymethyl cellulose, sorbitol and/or dextran. The suspension may also contain stabilizers.
The compositions as described herein may additionally contain other adjunct components conventionally found in pharmaceutical compositions. Thus, for example, the compositions may contain additional, compatible, pharmaceutically-active materials such as, for example, antipruritics, astringents, local anesthetics or anti-inflammatory agents, or may contain additional materials useful in physically formulating various dosage forms of the compositions of the present disclosure, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers. However, such materials, when added, should not unduly interfere with the biological activities of the components of the compositions of the present disclosure. The formulations can be sterilized and, if desired, mixed with auxiliary agents, including but not limited to, lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like which do not deleteriously interact with the antibody(s) of the formulation.
The isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, the vector or the pharmaceutical composition as described herein can be used in the treatment or prevention of diseases. Examples of such diseases include, but are not limited to, cancer and obesity. Thus, in one example, there is provided a method of treating or preventing, for example, but not limited to cancer and obesity, in a subject comprising administering an isolated cytotoxic peptide as described herein, an isolated nucleic acid molecule as described herein, a vector as described herein or a pharmaceutical composition as described herein.
In another example, there is provided an isolated cytotoxic peptide or conjugate thereof as described herein, an isolated nucleic acid molecule as described herein, a vector as described herein or a pharmaceutical composition as described herein, for use in treating or preventing, for example, but not limited to cancer and obesity, in a subject.
In yet another example, there is provided use of an isolated cytotoxic peptide or conjugate thereof as described herein, an isolated nucleic acid molecule as described herein or a vector as described herein in the manufacture of a medicament for treating or preventing, for example, but not limited to cancer and obesity, in a subject.
The major types of cancers that can be treated include but are not limited to carcinoma, sarcoma, lymphoma, germ cell tumor and blastoma. The specific types of cancers that can be treated include but are not limited to breast cancer, colorectal cancer, gastric cancer, melanoma, pancreatic cancer, skin cancer, leukemia, myeloma, hepatocellular cancer, pancreatic cancer, cervical cancer, ewings tumour, neuroblastoma, kaposis sarcoma, prostate cancer, bladder cancer, melanoma, lung cancer—non small cell lung cancer (NSCLC), and small cell lung cancer (SCLC), head and neck cancer, renal cancer, lymphoma, prostate cancer, neuroblastoma, a blood cancer, testicular cancer, ovarian cancer, liver cancer or esophageal cancer, cervical cancer, non-melanoma skin cancer, glioblastoma, carcinoma, uterus cancer, chronic lymphoid leukemia, lymphoblastic leukemia, follicular lymphomas, melanomas, malignant homeopathies, acute leukemia, basal cell carcinoma, bone cancer, brain and central nervous system (CNS) cancer, connective tissue cancer, eye cancer, kidney cancer, larynx cancer, Hodgkin's lymphoma, non-Hodgkin's lymphoma, oral cavity cancer (for example, lip, tongue, mouth, and pharynx), and rhabdomyosarcoma. In one specific example, the cancer is breast cancer.
In some examples, the cancer is invasive and/or metastatic cancer. In some other examples, the cancer is stage I cancer, stage II cancer, stage III cancer or stage IV cancer.
Stem cell therapy is a promising treatment for various diseases such as cancers, cardiovascular diseases, brain and spinal cord injury, blood related diseases, Parkinson's disease, Alzheimer's disease, diabetes, wound healing and so forth. The major types of stem cells include but are not limited to, embryonic stem cells, induced pluripotent stem cells and tissue stem cells. One main concern of the stem cell therapy, in particular the therapy using embryonic stem cells or induced pluripotent stem cells, is that the transplantation of any undifferentiated stem cells can lead to side effects such as the formation of teratoma, which is a tumor containing tissues derived from all three embryonic germ layers, i.e., ectoderm, mesoderm, and endoderm. To prevent the formation of teratoma resulted from stem cell transplantation, drugs with specific cytotoxic effect against undifferentiated stem cells can be used.
Thus, in one example, the isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, the vector or the pharmaceutical composition as described herein can be used in the prevention or treatment of diseases associated with stem cell transplantation. In some examples, the stem cell transplantation can be, but not limited to, transplantation of embryonic stem cells, transplantation of induced pluripotent stem cells, or transplantation of tissue stem cells, or a mixture thereof. One example of such diseases is teratoma. In some examples, the teratoma is benign, while in some other examples, the teratoma is malignant.
In another example, there is provided an isolated cytotoxic peptide or conjugate thereof as described herein, an isolated nucleic acid molecule as described herein, a vector as described herein or a pharmaceutical composition as described herein, for use in preventing or treating diseases associated with stem cell transplantation in a subject.
In yet another example, there is provided use of an isolated cytotoxic peptide or conjugate thereof as described herein, an isolated nucleic acid molecule as described herein or a vector as described herein in the manufacture of a medicament for preventing or treating diseases associated with stem cell transplantation in a subject.
In some examples, the subject that is being treated may be a mammal. In one specific example, the subject may be a human.
As used herein the terms “treatment”, “treating”, or other grammatical variants thereof, refers to any and all uses which remedy a disease state or symptoms, prevent the establishment of disease, or otherwise prevent, hinder, retard, or reverse the progression of disease or other undesirable symptoms in any way whatsoever. Treatments of cancer include but are not limited to: (i) the prevention or inhibition of cancer or cancer recurrence, (ii) the reduction or elimination of symptoms or cancer cells, and (iii) the substantial or complete elimination of the cancer in question. Treatment may be effected prophylactically (prior to the onset of the disease) or therapeutically (following diagnosis of the disease).
The isolated cytotoxic peptide or conjugate thereof, the isolated nucleic acid molecule, the vector or the composition as described herein may be provided in an amount that is therapeutically effective. The suitable amount includes a sufficient but non-toxic amount of the compound as described herein to provide the desired therapeutic effect. The exact amount required will vary from subject to subject depending on factors such as the species being treated, the age and general condition of the subject, the severity of the condition being treated, the particular agent being administered, the mode of administration, and so forth, and may be determined by one of ordinary skill in the art using only routine experimentation.
Dosing is dependent on severity and responsiveness of the disease state to be treated, with the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved. Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient. The administering physician can easily determine optimum dosages, dosing methodologies and repetition rates. Optimum dosages may vary depending on the relative potency of the composition. In general, dosage is from 0.01 μg to 100 g/kg of body weight, such as 0.01 g to 100 g, 0.05 μg to 90 g, 0.1 μg to 80 g, 0.5 μg to 70 g, 1 μg to 60 g, 10 μg to 50 g, 20 μg to 40 g, 30 μg to 30 g, 40 μg to 20 g, 50 μg to 10 g, 75 μg to 5 g, 100 μg to 4 g, 200 μg to 3 g, 300 μg to 2 g, 400 μg to 1 g, 500 μg to 900 mg, 600 μg to 800 mg, 700 μg to 700 mg, 800 μg to 600 mg, 900 μg to 500 mg, 1 mg to 400 mg, 2 mg to 300 mg, 3 mg to 200 mg, 3 mg to 100 mg, 4 mg to 90 mg, 5 mg to 80 mg, 6 mg to 70 mg, 7 mg to 60 mg, 8 mg to 50 mg, 9 mg to 40 mg, 10 mg to 30 mg or 15 mg to 25 mg/kg of body weight, and may be given once or more times daily, weekly, monthly or yearly. The treating physician can estimate repetition rates for dosing based on measured residence times and concentrations of the drug in bodily fluids or tissues. Following successful treatment, it may be desirable to have the subject undergo maintenance therapy to prevent the recurrence of the disease state, wherein the peptide or composition is administered in maintenance doses, ranging from 0.01 μg to 100 g/kg of body weight, once or more times daily, weekly, monthly, yearly, to once every 2 years.
The invention illustratively described herein may suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein. Thus, for example, the terms “comprising”, “including”, “containing”, etc. shall be read expansively and without limitation. Additionally, the terms and expressions employed herein have been used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention claimed. Thus, it should be understood that although the present invention has been specifically disclosed by preferred embodiments and optional features, modification and variation of the inventions embodied therein herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention.
The invention has been described broadly and generically herein. Each of the narrower species and subgeneric groupings falling within the generic disclosure also form part of the invention. This includes the generic description of the invention with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein.
Other embodiments are within the following claims and non-limiting examples. In addition, where features or aspects of the invention are described in terms of Markush groups, those skilled in the art will recognize that the invention is also thereby described in terms of any individual member or subgroup of members of the Markush group.
EXPERIMENTAL SECTION
Non-limiting examples of the invention will be further described in greater detail by reference to specific Examples, which should not be construed as in any way limiting the scope of the invention.
Example 1—HEXIM1 BR Peptide Induces Cell Death in a P-TEFb- and p53-Independent Fashion
HEXIM1 was originally identified from vascular smooth muscle cells treated with HMBA, an anti-proliferation compound. HEXIM1 is known as the inhibitor of P-TEFb. P-TEFb is a protein complex, which is composed of cyclin-dependent kinase 9 (CDK9) and a cyclin partner (cyclin T1, T2a, T2b, or K) with cyclin T1 being the predominant CDK9-associated cyclin. P-TEFb controls the elongation phase of RNAPII transcription and is required for transcriptional regulation of human immunodeficiency virus.
HEXIM1 exerts its inhibitory function on P-TEFb only when associated with the 7SK snRNA, while neither 7SK snRNA nor HEXIM1 alone instigates any effects. It is hypothesized that association of the 7SK snRNA with HEXIM1 leads to a conformational change that renders the cyclin T1-binding domain of HEXIM1 accessible for P-TEFb binding. HEXIM1 contains several functional domains and the domains required for 7SK snRNA and cyclin T1 binding have been identified. The N-terminus of HEXIM1, AA 1-150, has been characterized as a self-inhibitory domain (ID). Deletion of the ID enhances the inhibitory effects of HEXIM1 on P-TEFb activity. The region between AAs 150-180 of HEXIM1, which includes a stretch of basic residues, is referred to as the basic region (BR). The BR contains the binding motif for 7SK snRNA, KHRR (AAs 152-155). The P-TEFb binding motif, PYNT (AAs 202-205), is located between the BR and acidic region (AR, AAs 210-250). In the absence of 7SK snRNA, the AR can interact with the adjacent BR. Since the P-TEFb binding motif is located between the BR and AR, the BR-AR interaction may establish an auto-inhibitory conformation which prevents the association between HEXIM1 and P-TEFb. When 7SK snRNA binds to the BR, the BR-AR interaction is disrupted and the PYNT motif becomes accessible for P-TEFb binding. HEXIM1 can form a homodimer or a heterodimer with a HEXIM1-related protein, HEXIM2, through the dimerization domain (DD) at the C-terminus of HEXIM1.
When the 7SK snRNA binding motif, KHRR, is mutated to ILAA, the mutant HEXIM1 cannot interact with 7SK snRNA or regulate P-TEFb activity (FIG. 1B). It was found that FGF-BR-ILAA mutant still maintained its killing ability, in both HCT116 WT and p53 null cell lines, suggesting that P-TEFb might not be involved in the HEXIM1 BR-mediated cell killing (FIG. 1D).
HEXIM1 stabilizes p53 by blocking the HDM2-mediated ubiquitination of p53 and plays an essential role in p53 activation induced by anti-cancer drugs/compounds. HDM2, a p53-specific E3 ubiquitin ligase, is the master regulator of p53. Ubiquitination of p53 by HDM2 results in proteasome-mediated degradation of p53 protein. Six lysine residues, Lys-370, -372, -373, -381, -382, and -386, located within the C-terminal domain of p53 are known as the target residues for HDM2 ubiquitination. Six lysine residues located within the BR of HEXIM1 (Lys-150-152 and 159-161) are identified as the major sites for HDM2 ubiquitination. Sequence alignment of the ubiquitination sites between p53 (AAs 370-386) and HEXIM1 (AAs 150-161) exhibits similar distribution of the lysine residues (FIG. 1A). This suggests that HEXIM1 peptide containing these ubiquitination sites may have an impact on p53 activation, resulting in p53-dependent cell arrest or cell death.
A commercial p53 activating peptide, FGF-p53, containing the p53 ubiquitination sites (AAs 361-382) fused with a cell membrane-translocating peptide derived from Kaposi fibroblast growth factor, was reported to induce apoptosis in mutant and wild-type p53-bearing human cell lines. The internalized p53 peptide is likely to compete with the endogenous p53 protein in binding to HDM2 and protect the endogenous p53 from HDM2-mediated ubiquitination, resulting in stabilization and activation of p53. A FGF-BR fusion peptide was generated in the present disclosure, in which the cell penetrating FGF peptide was fused with HEXIM1 BR peptide containing the HDM2 ubiquitination residues (AAs 150-161). Two acute myeloid leukemia (AML) cell lines, AML2 and AML3, were treated with FGF-p53 and FGF-BR peptides and the effects of the peptides on cell viability were determined. FGF-p53 exhibited anti-proliferation effects on AML2 cells at high dosage (30 μM) but had no effects on AML3 cells (FIG. 11). The p53 level in AML2 has been shown to be significantly higher than that in AML3. This observation explained the p53-dependent anti-proliferation effects of FGF-p53 peptide in AML2 cells. However, FGF-BR peptide exhibited similar cytotoxicity on both cell lines and much stronger inhibition than FGF-p53 peptide, raising the possibility that the killing mediated by FGF-BR might not depend on p53 (FIG. 11).
Cytotoxicity was also observed in human cervical cancer cells, HeLa cells, when treated with FGF-BR peptide (FIG. 1C). Comparable cytotoxicity was also observed in normal cells, including HEK293 cells and human foreskin fibroblasts (HFFs) (FIG. 1C), suggesting the cytotoxic effect of BR peptide occurs to all cell types once it is internalized into cells.
The effect of FGF-BR peptide was also tested in human colon cancer cell line, HCT116 (p53 KO). The HCT116 (p53 WT) was used as a control. FGF-BR induced cell death in both HCT116 (p53 WT) and HCT116 (p53 KO) cell lines with similar potency, while the negative control peptide, FGF-X13, showed no effects in both cell lines (FIG. 1D). A fusion peptide, FGF-BR-RRR12 was generated, in which all six HDM2 ubiquitinating lysine residues were mutated to arginine and could no longer be ubiquitinated by HDM2 (FIG. 1B). If the BR peptide indeed induces cell death through the HDM2-p53 regulatory pathway, the BR-RRR12 mutant should lose its cytotoxic effects. FGF-BR-RRR12 did not exhibit reduced inhibitory effects but demonstrated stronger cell toxicity when compared to the wild-type FGF-BR peptide (FIG. 1D). It was also noticed that the p53 status had no effects on the cell killing mediated by FGF-BR-RRR12 (FIG. 1D), confirming that the cell death caused by the BR peptide was not p53-dependent.
Example 2—HEXIM1 BR Induced Cell Death is Independent of Apoptosis
The mechanism of cell death induced by the HEXIM1 BR peptide was examined. To monitor the real-time changes to the cells upon treatment with BR peptide, the effect of FGF-BR on HCT116 (p53 WT) and HCT116 (p53 KO) cells was examined in a live cell imaging setting. Cells treated with FGF peptide were included as a control. Within minutes, FGF-BR peptide rapidly induced drastic changes to the cell morphology with rupturing of the plasma membrane accompanied with damages to the nuclear membrane and abnormalities to the nucleolus in both cell types (FIG. 2A). No effects were detected when cells were treated with FGF peptide (FIG. 2A). Similar observations were seen in MCF7 breast cancer cells treated with LTV-tagged BR peptide, where BR peptide was conjugated to a breast cancer-targeting peptide, LTV (data not shown). Since BR-induced cytotoxicity occurred in minutes, whereas the duration of apoptosis is estimated to be from 12 to 24 hours, it is unlikely that FGF-BR lead to apoptosis. Moreover, the morphological changes seen in FGF-BR treated HCT116 cells do not resemble the characteristics of cells dying by apoptosis which include membrane blebbing and formation of apoptotic bodies. Cell swelling and subsequent rupturing of the plasma membrane followed by rapid lysis of the cells observed were descriptive of necrosis instead. In addition, the induction of cell death in LTV-BR-treated MCF7 cells could not be inhibited by a pan-caspase inhibitor z-VAD-Fmk (100 μM) (FIG. 2B), indicating that BR induced cell death is independent of apoptosis.
Taken together, it is unlikely that the conjugated FGF-BR peptide triggered apoptosis in the treated cells. Hence, using this BR peptide provides an attractive approach to eliminate cancer cells that have a defective apoptotic pathway.
Example 3—HEXIM1 BR Induces Rapid Depolarization of Mitochondrial Membrane Potential in a p53-Dependent Manner
It has been reported that a cytotoxic basic peptide, KLA, elicit its killing action by disrupting the mitochondrial membrane potential (MMP), which is important for ATP generation and induction of apoptosis. Since HEXIM1 BR contains many basic residues, it is possible that the BR may utilize similar mechanism for its potent killing. MMP was measured using JC-1 dye after incubating with FGF-X13 or FGF-BR peptides for three minutes. Carbonylcyanide m-chlorophenylhydrazone (CCCP), a known mitochondrial membrane depolarizer, was used as a positive control. FGF-BR treated HCT116 (p53 WT) cells experienced rapid mitochondrial depolarization within three minutes (FIG. 3A), which is similar to the time frame that morphological changes occurred upon addition of the FGF-BR peptide (FIG. 2A). However, no mitochondrial depolarization was observed in HCT116 (p53 KO) cells treated with FGF-BR (FIG. 3B), although these cells encountered rapid cytotoxicity similar to HCT116 (p53 WT) cells (FIG. 2A). It was known that p53 would translocate to the mitochondria, resulting in the reduction of MMP in p53-mediated apoptosis. Collectively, the data shows that HEXIM1 BR peptide is likely to induce cell killing though an alternative pathway that acts independently of p53 and apoptosis.
Example 4—HEXIM1 BR Peptide Alters Subcellular Localization of NPM and Reduces its Protein Expression
NPM is a multi-functional protein and participates in the process of ribosome biogenesis. In addition to its role in protein translation, NPM is required to maintain DNA integrity in cells. Knockout of NPM results in accumulation of DNA damage, which clearly indicates the essential role of NPM in cell proliferation and survival. NPM is located in nucleolus, the sub-cellular site of ribosome synthesis and assembly. About 35% of AML patients carry the cytoplasmic-misallocated mutant form of NPM, NPMc+. NPMc+ is found to interact and sequester a portion of HEXIM1 in the cytoplasm of the NPMc+ AML cell line and activates P-TEFb-dependent transcription, suggesting the involvement of HEXIM1 in tumorigenesis of AML. The BR domain of HEXIM1 was known to contain a nucleolar localization sequence. When the BR was fused with yellow fluorescent protein (YFP), the BR-YFP was found to localize in nucleoli. NPM was identified as a HEXIM1 binding protein, and the BR of HEXIM1 was required for NPM binding. To determine the effects of FGF-BR peptide on NPM, immunofluorescence was performed to examine the sub-cellular distribution of NPM in the FGF-BR-treated HCT116 (p53 WT) and HCT116 (p53 KO) cells. Normal nucleolar localization of NPM was observed in control experiments (FIG. 4A, DMSO and FGF-X13), while mislocalization of NPM was detected in both cell types when incubated with FGF-BR (FIG. 4A, FGF-BR). Furthermore, in both cell types, a reduction in NPM protein level was observed in the FGF-BR treated cells as compared to the FGF-X13 control (FIG. 4B). Various post-translational modifications of p53, such as phosphorylation and acetylation, have been shown to stabilize and activate p53 in response to cellular stress. The expression levels of phosphorylation of p53 on Ser15 and acetylation of p53 on Lys382 were investigated, and it was found that they remained unchanged in HCT116 (p53 WT) cells when treated with FGF-BR peptide (data not shown), suggesting a p53-independent pathway to trigger cell death. These results demonstrated that the BR peptide may interfere with protein translation/ribosome biosynthesis by disrupting sub-cellular localization of NPM and decreasing its expression, hence compromising its normal function.
Example 5—Selective Killing of LTV-BR Fusion Peptide on Breast Cancer Cells
Therapeutic peptides can be divided into two major categories: cell targeting and cytotoxic peptides. Targeting peptides, including the cancer targeting peptides, provide selectivity and specificity to direct the targeted treatment. Cytotoxic peptides can be used as the toxin/payload in fusion peptides, nano-particle conjugates, and antibody drug conjugates (ADCs) for potential cancer treatment.
To explore the potential use of the cytotoxic HEXIM1 BR peptide in cancer therapy, a breast cancer targeting peptide, LTV, was fused to HEXIM1 BR and BR-RRR12 to generate the fusion peptide, LTV-BR and LTV-BR-RRR12 (SEQ ID NO: 16). Both LTV-BR and LTV-BR-RRR12 peptides exhibited anti-proliferation effects in breast cancer cell lines MCF7 and MDA-MB-231, while no effects were detected using the LVT-X13 control peptide (FIGS. 5A and 5B). Although MDA-MB-231 cells are triple-negative with the absence of expression of oestrogen receptor, progesterone receptor and HER2, LTV-BR and LTV-BR-RRR12 elicited similar anti-cancer activity as compared to MCF7 cells (FIG. 5B). In addition, untagged control X13 (SEQ ID NO: 12) and the two BR peptides had no effect on cell viability (FIGS. 5C and 5D).
To define the active region of the BR peptide, a series of truncated BR peptides were generated based on the stretches of basic residues found in the BR peptide sequence. These truncated peptides were fused to LTV (SEQ ID Nos: 22-26) and introduced to MDA-MB-231 cells. The results suggest that the region encompassing the second and third stretch of basic residues (HRRRPSKKKRHW) is more critical in exerting cytotoxic activity as compared to the first stretch of basic residues (FIG. 7). To confirm this hypothesis, stretches of basic residues were replaced by alanine residues in the active region of BR (see SEQ ID Nos: 27-33). Results shown in FIG. 8 demonstrated the importance of the same stretches of basic residues (HRRRPSKKKRHW) in BR induced cytotoxicity. Nonetheless, all three stretches of basic residues are required to exhibit the maximum cytotoxic effect against MDA-MB-231 cells.
KLA peptide (SEQ ID NO: 13), a cytotoxic peptide, was often fused with a cancer-targeting peptide or conjugated to antibodies recognizing cancer cells (as antibody-drug conjugate, ADC) to exert cell-killing effect in anti-cancer therapy. LTV-KLA peptide (SEQ ID NO: 17) was generated and its effect was compared to that of LTV-BR. LTV-KLA exhibited stronger inhibition on the viability of MDA-MB-231 cells (FIG. 5B). However, it was noted that untagged KLA also showed non-specific killing on MDA-MB-231 cells, while BR- or BR-RRR12-treated cells remained highly viable (FIG. 5D). LTV-KLA also exhibited non-specific cell killing on non-breast cancer cell lines such as CHO (Chinese hamster ovary) and OPM-2 (multiple myeloma) cells, while little or no effects were observed in the LTV-BR-treated cells (FIGS. 9A and 9B). Similar observations were seen for normal human fibroblasts HFF and WI-38, indicating the specificity of LTV-BR towards breast cancer cell lines but not LTV-KLA (FIGS. 9C and 9D). It is predicted that a portion of the KLA ADCs will be broken down before reaching the target cancer cells, even though non-cleavable linkers may be used to generate these conjugates. As such, KLA peptides may be released from the conjugates, resulting in an off-target killing of normal cells/tissues. Taken together, the use of KLA as the toxic load in cancer therapy is questionable due to its non-specificity. In contrast, untagged HEXIM1 BR peptide is unable to kill cells when it is not fused with any cell penetrating or targeting peptides, suggesting that HEXIM1 BR peptide may be a safer alternative as compared to KLA, for the development of anti-cancer therapeutics.
Example 6—Cell Penetrating/Targeting Peptide is Required for Internalization HEXIM1 BR Peptide in Cells
The HEXIM1 BR peptide did not cause any cytotoxic effects when it was not fused with cell penetrating (for example, FGF) or cancer cell targeting (for example, LTV) peptides. It is thus possible that the unfused BR peptide may fail to internalize into cells without specific guidance. To test this hypothesis, fluorescent-labelled BR and LTV-BR peptides were generated and their presence in cells was examined by confocal microscope.
MCF7 cells were incubated with fluorescent-labeled peptides, followed by a washing step to remove the peptides failing to penetrate into cells. No fluorescent signals were observed in HEXIM1 BR peptide-treated cells (FIG. 6A), indicating that the BR peptide was not internalized. In the LTV-BR-incubated cells, fluorescent signals were detected within the cells (FIG. 6). LTV-BR was readily internalized into MCF7 cells and distributed in cytoplasm and nuclei (FIG. 6A). It was noted that its strong fluorescent signals were primarily localized in the nucleoli (FIG. 6A, LTV-BR-FITC). Detection of fluorescent signals in KLA-treated cells helps to explain the non-specific cytotoxicity induced by KLA peptide (FIG. 6A), while no fluorescent signal was observed in HEXIM1 BR-treated cells, indicating that the BR peptide could not enter the cells by itself (FIG. 6A, BR-FITC). Cells treated with LTV-KLA demonstrated that the sub-cellular localization of the peptide was observed mainly in the cytoplasm (FIG. 6A). The different distribution of LTV-BR and LTV-KLA suggests that BR and KLA may utilize different mechanisms for cell killing.
Flow cytometric analysis was also performed to quantify the amount of internalized fluorescent peptide in MCF7 cells. LTV peptide directed the uptake of almost 100% of LTV-fused peptides (LTV-BR and LTV-KLA) into the breast cancer cell line (FIG. 6B). Approximately 65% of KLA-FITC-treated MCF7 cells internalized KLA-FITC, whereas there was no entry of BR-FITC into MCF7 cells (FIG. 6B). These results clearly demonstrate the safety feature of HEXIM1 BR peptide when compared to the non-specific cytotoxic KLA peptide. LTV assisted in the cellular internalization of HEXIM1 BR peptide into its target cells and the nucleolar localization of the fusion peptide might be subsequently guided by BR peptide.
Example 7—Application of HEXIM1 BR Peptide in Weight Loss Study
2.5×104 cells/ml of mouse fibroblast cells 3T3 were cultured in gelatin coated plates in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (Gibco) to reach 100% confluent. The cells were then maintained at 100% confluency by supplementing with fresh media in a 37° C. incubator with 5% CO2 for 3 days. For induction, cells were then cultured in DMI Induction Media (which is DMEM supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin, 0.1% 3-isobutyl-1-methylxanthine (sigma), 0.1% insulin (sigma) and 0.1% dexamethasone (sigma)) for 48 hours. The cells were then cultured in Insulin Induction Media (which is DMEM supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin and 0.1% insulin). The insulin induction media was changed every 2 days until the cells were differentiated to adipose cells.
A D-form BR peptide with the sequence of QLGKKKHRRRPSKKKRHW was tagged to a fat tissue targeting peptide of the sequence CKGGRAKDC (ATS peptide, SEQ ID NO: 39) to generate the ATS-BRD peptide of the sequence CKGGRAKDCGG(QLGKKKHRRRPSKKKRHW)D (SEQ ID NO: 40). Both undifferentiated 3T3 cells and differentiated adipose cells were treated with the ATS-BRD peptide. ATS-tagged KLAD peptide was used as a positive control. Cell viability results shown in FIG. 10 indicates that the ATS-BRD peptide has no cytotoxic effect on undifferentiated 3T3 mouse fibroblast cells, but has significant cytotoxic effect on differentiated adipose cells. The cytotoxic effect of ATS-BRD peptide on adipose cells is of similar potency as ATS-KLAD peptide.
Example 8—Use of HEXIM1 BR Peptide as the Toxin for Antibody Drug Conjugates (ADCs)
To generate ADCs, the cytotoxic HEXIM1 BR peptides are covalently attached to antibodies through chemical linkers. Cysteine and lysine are two most naturally occurring amino acids which are used to attach the toxin through the linker to the antibody. To conjugate at the lysine residues of antibodies, two chemical linkers, N-succinidyl-3-(2-pyridylothio)propionate (SPDP) and succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC), are utilized according to manufacturer's manual (Thermo Fisher Scientific). Briefly, the linkers are first crosslinked to the lysine residues of the antibodies. As the thiol group is required for conjugating the peptides to the chemical linkers, a cysteine residue is added at the N- or C-terminal of the BR peptide. After removing excess linkers and peptides, the effects of the BR-conjugated ADCs are analyzed in vitro using the selected breast cancer cell lines.
IgGs have four pairs of interchain disulfide bonds, two between the heavy chains in the hinge region and two on Fab between heavy and light chains. Intrachain disulfide bonds are to be left intact because they are critical to maintain the basic IgG domain structure essential for antigen recognition. Only partial reduction of the interchain disulfide bonds at the hinges gives eight potential conjugation sites through cysteine residues.
Example 9—Materials and Methods
Cell Lines—
Human cell lines including HeLa, HEK293, MCF7, MDA-MB-231, CHO-K1, OPM-2, and WI-38 were obtained from American Type Culture Collection. AML2 and AML3 cells were purchased from Deutsche Sammlung von Mikroorganismen und Zellkulturen. HCT116 p53+/+ and p53−/− cells were kindly given by Dr. Bert Vogelstein. Primary human foreskin fibroblasts (HFF) were obtained from Dr. Mark Stinski. HCT116, HCT116 (p53 KO), HeLa, 293, HFF, MCF7, CHO-K1, and WI-38 cells were cultured in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (Gibco). MDA-MB-231 and OPM-2 cells were cultured in RPMI-1640 (Gibco) containing 10% FBS and 1% penicillin/streptomycin. All cells were routinely maintained in a 37° C. incubator with 5% CO2.
Peptide Synthesis—
All peptides used in this study were chemically synthesized and purified by high performance liquid chromatography with >98% purity (First Base, Singapore). Their sequences are available in the Supplementary Table 1. Stock solutions were obtained by reconstituting the powder in sterile water or 50% DMSO and stored at −80° C.
Immunoblotting Analysis—
Cells were lysed in lysis buffer [50 mM Tris-HCl, (pH 7.5), 150 mM NaCl, 1% NP40, 0.5% sodium deoxycholate, Protease Inhibitor tablet (Roche)] and used for western blotting. Western blotting was carried out as previously mentioned. The primary antibodies used include anti-NPM (Invitrogen) and anti-actin (Millipore). The film of western blot was scanned, and the protein bands were quantified by the GS-800 densitometer (Bio-Rad). The protein level of NPM was quantified after normalizing with the loading control, actin.
Cell Viability and Cytotoxicity Assays—
Cells were plated in clear-bottomed white walled 96-well plates (Corning) and incubated overnight. Cells were treated with indicated peptides in 1% FBS-containing media for overnight or indicated timings at 37° C. Cell viability was measured with CellTiter-Glo reagent (Promega) according to the manufacturer's instructions. For cytotoxicity assay, cells were plated in black walled 96-well plates (Corning) to allow them to adhere overnight. Upon treatment with a pan-caspase inhibitor, z-VAD-Fmk (100 M) (Sigma) and subsequent treatment with LTV-tagged peptides, cytotoxicity induced was determined by CellTox cytotoxicity assay (Promega) 30 minutes after addition of peptides according to the manufacturer's instructions. Luminescence was determined using an Infinite 200 multiplate reader (Tecan).
Measurement of MMP—
The cationic fluorescent dye 1,1′,3,3′-tetraethylbenzamidazolocarbocyanin iodide (JC-1) (Invitrogen) was utilized for MMP measurement. JC-1 was dissolved in DMSO (200 μM). Peptide-treated cells in 96-well deep sided, clear bottom, dark sided microplates were incubated with media containing JC-1 for 30 min at 37° C. and then washed twice with warm PBS. Changes in MMP were determined using a multiplate spectrofluorometer (Tecan) (excitation: 475 nm; emission: 530 nm (green); emission: 590 nm (red)). The decrease in the ratio of red to green fluorescence was used to determine relative mitochondrial depolarization.
Flow Cytometry—
MCF7 cells were plated on 6-cm culture dish to allow overnight adherence. FITC-labeled peptides (30 μM) were added to the cells, incubated for 30 min at 37° C., and subsequently washed three times with PBS. The cells were then trypsinized, collected by centrifugation, and finally resuspended in 500 μl ice-cold 2% FBS-containing PBS for flow cytometry analysis.
Immunofluorescence and Confocal Microscopy—
For staining with anti-NPM, cultured cells were fixed in 10% neutral buffered formalin (Sigma) for 10 min, and then in methanol for 10 min, washed in PBS and incubated with blocking buffer (PBS containing 0.5% bovine serum albumin) for 1 hour prior to incubation with a mouse anti-NPM antibody (Invitrogen) in blocking buffer for overnight at 4° C. Cells were then incubated with Alexa Fluor 488-conjugated secondary antibody (Jackson Immuno Research Laboratories) for 1 hour, washed three times with PBS and counterstained with 4′,6-diamidino-2-phenylindole (DAPI)-containing mounting solution (Vectashield). Stained cells were examined with a LSM 510 confocal microscope using a 63× objective lens (Zeiss).
To determine the ability of FITC-labeled peptides to enter the cells and to visualize intracellular distribution of the peptides, MCF7 cells were plated on 4-chamber glass cover slides (Lab-Tek) to adhere overnight, incubated with FITC-labeled peptides (30 μM) for 30 min, and then washed three times with PBS before being fixed and mounted with DAPI-containing mounting solution (Vectashield). Images were acquired using a Nikon AIR confocal laser scanning microscope equipped with a 60× oil-immersion objective lens (SBIC-Nikon Imaging Centre).
Statistical Analysis—
All experiments were performed independently for at least three times. All statistical analyses for comparison between two groups were performed with two-tailed unpaired student's t-test using the Prism 5.01 (GraphPad Software).

Claims (9)

What is claimed is:
1. A recombinant peptide comprising a cytotoxic peptide and an internalizing peptide, wherein the cytotoxic peptide is conjugated to the internalizing peptide, and wherein the cytotoxic peptide is represented by the consensus sequence Q1-L2-G3-X4-X5-X6-X7-X8-X9-R10-P11-S12-X13-X14-X15-R16-H17-W18 (SEQ ID NO: 36),
wherein,
i) each of X4, X5, and X6 is any amino acid;
ii) X7 is histidine or leucine;
iii) X8 and X9 are independently K or R or H or A; and
iv) X13, X14 and X15 are independently K or R or H;
and, wherein the internalizing peptide is a cancer cell-targeting peptide selected from the group consisting of arginine-glycine-aspartic acid (RGD), asparagine-glycine-arginine (NGR), TCP-1 phage peptide (TCP-1) and any one of the peptides of SEQ ID Nos: 8-11, 64-789 and 795-798.
2. The recombinant peptide of claim 1, wherein
v) X8 and X9 are the same, and are K or R or H or A; and
vi) X13, X14 and X15 are the same, and are K or R or H.
3. The recombinant peptide of claim 1, wherein the amino acid sequence of the cytotoxic peptide is selected from the group consisting of: i) QLGKKKHRRRPSKKKRHW (SEQ ID NO: 3), ii) QLGRRRHRRRPSRRRRHW (SEQ ID NO: 4), iii) QLGKKILAARPSKKKRHW (SEQ ID NO: 5), and iv) QLGAAAHRRRPSKKKRHW (SEQ ID NO: 38).
4. A peptide conjugate comprising a cytotoxic peptide and a nanoparticle or a microparticle, wherein the cytotoxic peptide is conjugated to the nanoparticle or microparticle, and wherein the cytotoxic peptide is represented by the consensus sequence Q1-L2-G3-X4-X5-X6-X7-X8-X9-R10-P11-S12-X13-X14-X15-R16-H17-W18 (SEQ ID NO: 36),
wherein,
i) each of X4, X5, and X6 is any amino acid;
ii) X7 is histidine or leucine;
iii) X8 and X9 are independently K or R or H or A; and
iv) X13, X14 and X15 are independently K or R or H.
5. The peptide conjugate of claim 4, wherein
v) X8 and X9 are the same, and are K or R or H or A; and
vi) X13, X14 and X15 are the same, and are K or R or H.
6. The peptide conjugate of claim 4, wherein the amino acid sequence of the cytotoxic peptide is selected from the group consisting of: i) QLGKKKHRRRPSKKKRHW (SEQ ID NO: 3), ii) QLGRRRHRRRPSRRRRHW (SEQ ID NO: 4), iii) QLGKKILAARPSKKKRHW (SEQ ID NO: 5), and iv) QLGAAAHRRRPSKKKRHW (SEQ ID NO: 38).
7. The peptide conjugate of claim 4, wherein the nanoparticle or microparticle is made from a material selected from the group consisting of metal, silica, carbon, polymeric materials, and mixtures thereof.
8. The recombinant peptide of claim 1, wherein the peptide has a modification selected from the group consisting of glycosylation, sulfation, phosphorylation, ubiquitination, methylation, lipidation, biotinylation, hydroxylation and acetylation.
9. The recombinant peptide of claim 1, comprising the peptide of SEQ ID NO: 15.
US15/555,482 2015-03-04 2016-03-03 Cytotoxic HEXIM1 peptides and uses thereof Expired - Fee Related US10500288B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
SG10201501642V 2015-03-04
SG10201501642V 2015-03-04
SG10201504803T 2015-06-17
SG10201504803T 2015-06-17
PCT/SG2016/050101 WO2016140624A1 (en) 2015-03-04 2016-03-03 Cytotoxic hexim1 peptides and uses thereof

Publications (2)

Publication Number Publication Date
US20180243441A1 US20180243441A1 (en) 2018-08-30
US10500288B2 true US10500288B2 (en) 2019-12-10

Family

ID=56848975

Family Applications (1)

Application Number Title Priority Date Filing Date
US15/555,482 Expired - Fee Related US10500288B2 (en) 2015-03-04 2016-03-03 Cytotoxic HEXIM1 peptides and uses thereof

Country Status (6)

Country Link
US (1) US10500288B2 (en)
EP (1) EP3265472A4 (en)
CN (1) CN107531757A (en)
HK (1) HK1248729A1 (en)
SG (1) SG11201705759SA (en)
WO (1) WO2016140624A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190321496A1 (en) * 2016-06-16 2019-10-24 Baker Idi Heart And Diabetes Institute Holdings Limited Agents and methods for the diagnosis and treatment of diseases associated with extracellular matrix turnover
JP2020528739A (en) * 2017-06-28 2020-10-01 ユニヴァーシティ オブ サウス フロリダ Modified UBE3A gene for gene therapy for Angelman syndrome
WO2020085767A1 (en) * 2018-10-22 2020-04-30 한양대학교 산학협력단 Cancer cell death composition and use thereof
KR102246627B1 (en) * 2019-08-27 2021-04-29 재단법인대구경북과학기술원 A composition comprising hmba for preventing and treating obesity
CN112521446B (en) * 2020-11-06 2022-06-21 浙江海洋大学 ACE inhibitory peptide and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6753418B2 (en) * 2000-10-05 2004-06-22 Case Western Reserve University Suppressors of human breast cancer cell growth
WO2012129395A2 (en) 2011-03-23 2012-09-27 The Research Foundation Of State University Of New York Diagnosis and treatment of prostate cancer
US8470976B2 (en) * 2006-06-17 2013-06-25 Board Of Regents, The University Of Texas System Methods and compositions for targeting macromolecules into the nucleus

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6753418B2 (en) * 2000-10-05 2004-06-22 Case Western Reserve University Suppressors of human breast cancer cell growth
US8470976B2 (en) * 2006-06-17 2013-06-25 Board Of Regents, The University Of Texas System Methods and compositions for targeting macromolecules into the nucleus
WO2012129395A2 (en) 2011-03-23 2012-09-27 The Research Foundation Of State University Of New York Diagnosis and treatment of prostate cancer

Non-Patent Citations (58)

* Cited by examiner, † Cited by third party
Title
A chromatin Landmark and Transcription Initiation at Most Promoters in Human Cells, by Guenther MG, Levine SS, Boyer LA, Jaenisch R, Young RA; Cell 130, 77-88; 2007; pp. 77-88.
A mitochondria-Trageting Gold-Peptide Nanoassembly for Enhanced Cancer-Cell Killing, by Ma X, Wang X, Zhou M, Fei H; Advanced Healthcare Materials; 2013; pp. 1638-1643.
A Proapoptotic Peptide for the Treatment of Solid Tumors, by Mai JC, Mi Z, Kim SH, Ng B, Robbins PD; Cancer Research; American Association for Cancer Research 61; 2001; pp. 7709-7712.
A Proapototic Peptide Conjugated to Penetratin Selectively Inhibits Tumor Cell Growth, by Alves ID, Carré M, Montero MP, Castano S, Lecomte S, Marquant R, Lecorché P, Burlina F, Schatz C, Sagan S, Chassaing G, Braguer D, Lavielle S; Biochimica et Biophysica Acta; 2014; pp. 2087-2098.
Anti-cancer activity of targeted pro-apoptotic peptides, by H. M Ellerby, W Arap, L M Ellerby, R Kain, R Andrusiak, G Del Rio, S Krajewski, C R Lombardo, R Rao, E Ruoslahti, D E Bredesen & R Pasqualini; Nature America Inc.; vol. 5, No. 9; Sep. 1999; pp. 1032-1038 < https://www.medicine.nature.com/>.
Apoptosis Inducing, Conformationally Constrained, Dimeric Peptide Analogs of KLA with submicromolar cell penetrating abilities, by Hyun S, Lee S, Kim S, Jang S, Yu J, Lee Y; Biomacromolecules; ACS Publications 2014; pp. 3746-3752.
Binding of the 7SK snRNA turns the HEXIM1 protein into a P-TEFb (CDK9/cyclin T) inhibitor, by Michels AA, Fraldi A, Li Q, Adamson TE, Bonnet F, Nguyen VT, Sedore SC, Price JP, Price DH, Lania L, Bensaude O; European Molecular Biology Organization; 2004; pp. 2608-2619.
Brd4 and HEXIM1: Multiple Roles in P-TEFb Regulation and Cancer, by Ruichuan Chen, Jasper H. N. Yik, Qiao Jing Lew, and Sheng-Hao Chao; BioMed Research International; vol. 2014, Article ID 232870, 11 pages.
Bryan Whitmann, et al., "The breast cell growth inhibitor, estrogen down regulated gene 1, modulates a novel functional interaction between estrogen receptor alpha and transcriptional elongation factor cyclin T1," Oncogene, vol. 24, No. 36, pp. 5576-5586 (May 23, 2005).
Bryan Whittmann, et al., "Identification of a novel inhibitor of breast cell growth that is down-regulated by estrogens and decreased in breast tumors," Cancer Res., vol. 63, No. 16, pp. 5151-5158 (Aug. 15, 2003).
Cancer Research; Amphipathic peptide-based Fusion Peptides and Immunoconjugates for the Targeted Ablation of Prostate Cancer Cells, by Kaushal Rege, Suraj J. Patel, Zaki Megeed and Martin L. Yarmush; American Association for Cancer Research; 2007; pp. 6368-6375.
Cancer Research; Selective Apoptotic killing of Malignant Hemopoietic Cells by Anitibody-Targeted Delivery of an Amphipathic Peptide, by A J. Marks, M S. Cooper, R J. Anderson, K H. Orchard, G Hale, J M. North, K Ganeshaguru, A J. Steele, A B. Mehta, M W. Lowdell & R. G Wickremasinghe; American Association for Cancer Research; 2005; 65: pp. 2373-2377.
Cancer statistics, 2014, by Siegel R, Ma J, Zou Z, Jernal A.; CA Cancer J Clin; Jan.-Feb. 2014; vol. 64, No. 1; pp. 9-29.
Cationic amphiphilic peptides with cancer-selective toxicity, by F Schweizer; European Journal of Pharmacology 625; 2009; pp. 190-194.
Cloning of hexamethylene-bis-acetamide-inducible transcript, HEXIM1, in human vascular smooth muscle cells, by M Kusuhara, K Nagasaki, K Kimura, NMaass, T Manabe, S Ishikawa, M Aikawa, K Miyazaki, K Yamaguchi; Biomedical Research 20 (5); 1999; pp. 273-279.
Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype, by F B, Mecucci C, Tiacci E, A M, RR, Pasqualucci L, La Starza R, Diverio D, Colombo E, Santucci A, Bigerna B, Pacini R, Pucciarini A, et al.; The New England Journal of Medicine; 2007; pp. 254-267.
Dual-Targeting Pro-apototic Peptide for Programmed cancer cell death via specific mitochondria damage, by Chen WH, Xu XD, Luo GF, Jia HZ, Lei Q, Cheng SX, Zhuo RX, Zhang XZ; Scientific Reports 3:3468; pp. 1-7.
Expression of Bcl-xl Can Confer a Multidrug Resistance Phenotype by Minn AJ, Rudin CM, Boise LH, Thompson CB; Blood Journal, vol. 86, No. 5; 2017; pp. 1903-1910.
Flavopiridol Inactives P-TEFb and Blocks Most RNA Polymerase II Transcription in Vivo*, by Chao SH1, Price DH; The Journal of Biological Chemistry; American Society for Biochemistry and Molecular Biology, Inc.; 2001; pp. 31793-31799.
Flavopiridol Inhibits P-TEFb and Blocks HIV-1 Replication*, by Chao SH, Fujinaga K, Marion JE, Taube R, Sausville EA, Senderowicz AM, Peterlin BM, Price DH; The Journal of Biological Chemistry; vol. 275, No. 37; 2000; pp. 28345-28348.
HEIM1 Induces Differentiation of Human Pluripotent Stem Cells, by V Ding, Q J Lew, K L Chu, S Natarajan, V Rajasegaran, M Gurumurthy, A B. H. Choo, Sheng-Hao Chao; Open Access; PLOS One; 2013; <https://doi.org/10.1371/journal.pone.0072823>.
HEMIM1 and the Control of Transcription Elongation From Cancer and Inflammation to AIDS and Cardiac Hypertrophy, by Dey A, Chao SH, Lane DP; Landes Bioscience; Cell Cycle 6:15; vol. 6, Issue 15; 2007; pp. 1856-1863.
HEXIM1, a New Player in the P53 Pathway, by Lew QJ, Chu KL, Chia YL, Cheong N, Chao SH; Cancers (Basel). 2013; pp. 838-356 <www.mdpi.com/journals/cancers>.
Human Cytomegalovirus: Glycoproteins Associated with Virions and Dense Bodies, by Mark F. Stinski; American Society for Microbiology; Journal of Virology vol. 19, No. 2; 1976; pp. 594-609.
Identification of a prominent nuclear protein associated with proliferation of normal and malignant B Cells, by Feuerstein N, Mond JJ; The Journal of Immunology 139; 1987; pp. 1818-1822.
Inhibition of metastasis by HEXIM1 through effects on cell invasion and angiogenesis, by Ketchart W, Smith KM, Krupka T, Wittmann BM, Hu Y, Rayman PA, Doughman YQ, Albert JM, Bai X, Finke JH, Xu Y, Exner AA, Montano MM; National Institute of Health (NIH) Public Access, Author Manuscript, Oncogene 2013; 26 pages.
Inhibition of P-TEFb (CDK9/Cyclin T) kinase and RNA polymerase II transcription by the coordinated actions of HEXIM1 and 7SK snRNA, by Yik JH1, Chen R, Nishimura R, Jennings JL, Link AJ, Zhou Q; Molecular Cell vol. 12; Oct. 2003; pp. 971-982.
Interplay between 7SK snRNA and oppositely charged Regions in HEXIM1 Direct the inhibition of P-TEFb, by Matjaz barboric, Jiri Kohoutek, Jason P Price, Dalibor Blazek, David H Price & B Matija Peterlin; The European Molecular Biology Organization (EMBO) Journal; 2005; pp. 4291-4303.
Joanne Lau, et al., "Ubiquitination of HEXIM1 by HDM2," Cell Cycle, Vo. 8, No. 14, pp. 2247-2254 (Jul. 15, 2009).
Kinetics of Apoptotic Markers in Exogeneously Induced Apoptosis of EL4 Cells, by Jessel R, Haertel S, Socaciu C, Tykhonova S, Diehl HA; J Cell Mol Med, vol. 6, No. 1; 2002; pp. 82-92.
Mapping the functional domains of nucleolar protein B23, by Hingorani K, Szebeni A, Olson MO; The Journal of Biological Chemistry; vol. 275, No. 32; 2000; pp. 24451-24457.
Meera Gurumurthy, et al., "Nucleophosmin Interacts with HEXIM1 and Regulates RNA Polymerase II Transcription," J Mol Biol., vol. 378, No. 2, pp. 302-317 (Mar. 4, 2008).
Morphologic Criteria and Detection of Apoptosis, by Antti Saraste; Hertz; Urban & Vogel; 1999; pp. 189-195.
Multiple C-Terminal Lysine Residues Target p53 for Ubiquitin-Proteasome-Mediated Degradation, by Manuel S. Rodriguez, Joana M. P. Desterro, Sonia Lain, David P. Lane, and Ronald T. Hay; Molecular and Cellular Biology, vol. 20, No. 22; American Society for Microbiology; 2000; pp. 8458-8467.
Necrosis Methods and Protocols, by Kimberly McCall, & Charles Klein; Methods in Molecular Biology, Springer Protocols; Humana Press; 2013; 253 Pages.
Necrosis: A specific Form of Programmed Cell Death?, by Proskuryakov SY, Konoplyannikov AG, Gabai VL; Science Direct, Experimental Cell Research 283; 2003; pp. 1-16.
Necrotic Death as a Cell Fate, by Zong WX & Thompson CB; Genes & Development 20; Cold Spring Harbor Laboratory Press; 2006; pp. 1-15.
Noritada Yoshikawa, et al., "Cardiomyocyte-Specific Overexpression of HEXIM1 Prevents Right Ventricular Hypertrophy in Hypoxia-Induced Pulmonary Hypertension in Mice," PloS One, vol. 7, No. 12, pp. 1-13 (Dec. 31, 2012).
NPMc+ AML cell line shows differential protein expression and lower sensitivity to DNA-damaging and p53-inducing anticancer compounds, by Qiao Jing Lew,Chuan Hao Tan,Meera Gurumurthy,Kai Ling Chu,Nge Cheong,David P. Lane &Sheng-Hao Chao; Cell Cycle 10:12; Landes Bioscience; Jun. 2011; pp. 1978-1987.
Nucleophosmin and cancer, by Silvia Grisendi, Cristina Mecucci, Brunangelo Falini & Pier Paolo Pandolfi; Nature Reviews Cancer, vol. 6; 2006; pp. 493-505.
Overexpression of bcl-2 Protects Prostate Cancer Cells from Apoptosis in Vitro and Confers Resistance to Androgen Depletion in Vivo, by Raffo AJ, Perlman H, Chen MW, Day ML, Streitman JS, Buttyan R; Canccer Research 55; 1995; pp. 4438-4-445.
PCT International Search Report for PCT Counterpart Application No. PCT/SG2016/050101, 6 pgs. dated (May 13, 2016).
PCT Written Opinion for PCT Counterpart Application No. PCT/SG2016/050101, 6 pgs. dated (May 13, 2016).
Peptides Targeting Caspase Inhibitors*, by Tamm I, Trepel M, Cardó-Vila M, Sun Y, Welsh K, Cabezas E, Swatterthwait A, Arap W, Reed JC, Pasqualini R; The Journal of Biological Chemistry; American Society for Biochemistry and Molecular Biology, Inc., vol. 278, No. 16; 2003; pp. 14401-14405.
Polycomb Protien EZH2 Supresses Apoptosis by silencing the proapoptotic miR-31, by Zhang Q, Padi SK1, Tindall DJ2, Guo B; Cell Death Disease; NPG; 2014; 7 Pages.
P-TEFb, a Cyclin-Dependent Kinase Controlling Elongation by RNA Polymerase II, by David H. Price; Molecular and Cellular Biology; American Society for Microbiology; vol. 20, No. 8; 2000; pp. 2629-2634.
Qiao Jing Lew, et al., "Identification of HEXIM1 as a Positive Regulator of p53," J. Biol Chem, vol. 287, No. 43, pp. 36443-36454 (Sep. 4, 2012).
Requirement for p53 and p21 to Sustain G2 Arrest After DNA Damage, by Bunz F, Dutriaux A, Lengauer C, Waldman T, Zhou S, Brown JP, Sedivy JM, Kinzler KW, Vogelstein B; Reports; Science Magazine vol. 282; 1998; pp. 1497-1501.
Restoration of the Growth Suppression Function of mutant p53 by a Synthetic Peptide Derived from the p53 C-terminal Domain, by Selivanova G, Iotsova V, Okan I, Fritsche M, Ström M, Groner B, Grafström RC, Wiman KG; Nature Medicine, vol. 3, No. 6; 1997; pp. 632-638.
Selective killing of cancer cells by peptide-targeted delivery of an anti-microbial peptide, by Sioud M, Mobergslien A; Biochemical Pharmacology 84; 2012; pp. 1123-1132.
Shu Hui Neo, et al., "Use of a novel cytotoxic HEXIM1 peptide in the directed breast cancer therapy," Oncotarget, vol. 7, No. 5, pp. 5483-5494 (Dec. 29, 2015).
Sioud, Biochemical Pharmacology 84 (2012) 1123-1132 (Year: 2012). *
Studies on mechanism of action of anticancer peptides by modulation of hydrophobicity within a defined structural framework, by Huang YB, Wang XF, Wang HY, Liu Y, Chen Y; Therapeutic discovery; Molecular Cancer Therapeutics; American Association for Cancer Research; 2011; pp. 416-427.
Synthetic therapeutic peptides: science and market, by Vlieghe P, Lisowski V, Martinez J, Khrestchatisky M; Drug Discovery Today, vol. 15, Nos. ½; Jan. 2010; pp. 40-56.
Targeted Nanoparticle enhanced Proapoptotic peptide as potential therapy for glioblastoma, by Agemy L, Friedmann-Morvinski D, Kotamraju VR, Roth L, Sugahara KN, Girard OM, Mattrey RF, Verma IM, Ruoslahti E; Proc Natl Acad Sci (PNAS) vol. 108, No. 42; 2011; pp. 17450-17455.
Targeting the Na+/K+-ATPase α1 Sbunit of Hepatoma HepG2 Cell Line to Induce Apoptosis and Cell Cycle Arresting, by Xu ZW1, Wang FM, Gao MJ, Chen XY, Hu WL, Xu RC; Biol Pharm Bull, vol. 33, No. 5; Pharmaceutical Society of Japan; 2010; pp. 743-751.
The 3′UTR of HIC mRNA improves the production of recombinant proteins in Chinese hamster ovary cells, by Liu J, Ku SC, Lee J, Young TM, Pe'ery T, Mathews MB, Chao SH; Journal of Biotechnology 139; 2009; pp. 152-155.
The Design, Synthesis, and Evaluation of Molecules that Enable or Enhance Cellular Uptake: Peptoid Molecular Transporters, by Wender PA, Mitchell DJ, Pattabiraman K, Pelkey ET, Steinman L, Rothbard JB; Proc Natl Acad Sci, vol. 97, No. 24; 2000; pp. 13003-13008.

Also Published As

Publication number Publication date
EP3265472A1 (en) 2018-01-10
US20180243441A1 (en) 2018-08-30
CN107531757A (en) 2018-01-02
SG11201705759SA (en) 2017-08-30
HK1248729A1 (en) 2018-10-19
EP3265472A4 (en) 2018-07-25
WO2016140624A1 (en) 2016-09-09

Similar Documents

Publication Publication Date Title
US10500288B2 (en) Cytotoxic HEXIM1 peptides and uses thereof
EP2418217B1 (en) Cell-permeable peptide inhibitors of the JNK signal transduction pathway
KR101799904B1 (en) Cell-penetrating peptide, conjugate comprising same and composition comprising same
US7943581B2 (en) Cell penetrating peptides for intracellular delivery of molecules
CA2632451C (en) Cell penetrating peptides for intracellular delivery of molecules
CN109513010B (en) Self-assembling drug nanoconjugates for tumor cell specific responses
US9617344B2 (en) Fusion protein comprising targeting moiety, cleavage site, and cell membrane penetrating domain, and use thereof
KR102301596B1 (en) Multi-ligand drug conjugates and uses thereof
CN112533640A (en) Targeting of tumor-associated macrophages of type M2 using melittin-based apoptosis-inducing peptides
EP2044125B1 (en) Compositions and methods for delivering anti-activated ras antibodies into cells
EP1795539B1 (en) Cell penetrating peptides for intracellular delivery of molecules
KR101456026B1 (en) Peptide Having Turmor Selective Permeability and Use Thereof
EP3160489B9 (en) Cell-permeable peptide inhibitors of the jnk signal transduction pathway for the treatment of cystitis
Huang et al. Cloning, expression, purification and functional characterization of the oligomerization domain of Bcr–Abl oncoprotein fused to the cytoplasmic transduction peptide
US11576978B2 (en) Hemoglobin-based therapeutic agents
US20240052005A1 (en) Improved cell-penetrating peptides and fusion proteins
AU2021274149B2 (en) Novel nucleolin-binding peptide and use thereof
JP2024517518A (en) Macrophage targeting peptides, and conjugates, compositions and uses thereof
CA3153060A1 (en) Peptides targeting macrophages, and conjugates, compositions, and uses thereof
KR20230144960A (en) Immunotherapy drug based on new peptide
WO2022234346A1 (en) Peptides targeting macrophages, and conjugates, compositions, and uses thereof

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

AS Assignment

Owner name: AGENCY FOR SCIENCE, TECHNOLOGY AND RESEARCH, SINGA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CHAO, SHENG-HAO;REEL/FRAME:043631/0714

Effective date: 20160404

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20231210